1
|
Møller MW, Nortvig MJ, Andersen MS, Poulsen FR. DNA Methylation in Pituitary Adenomas: A Scoping Review. Int J Mol Sci 2025; 26:531. [PMID: 39859246 PMCID: PMC11765255 DOI: 10.3390/ijms26020531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Pituitary adenomas are a diverse group of neoplasms with variable clinical behavior. Despite advances in genetic analysis, understanding the role of epigenetic modifications, particularly DNA methylation, remains an area under investigation. This scoping review aimed to update and synthesize the current body of literature on DNA methylation in pituitary adenomas, focusing on methodological advancements and clinical correlations. A systematic search conducted across multiple databases, including Embase, Scopus, MEDLINE, and CENTRAL, identified 107 eligible studies. Early methods, such as methylation-restricted digestion and methylation-specific PCR (MSP), have evolved into more comprehensive approaches, such as chip-based DNA methylation analysis. Key findings suggest that genes like POMC, SOCS-1, and RASSF1A show a significant association between methylation and clinical behavior. However, methylation patterns alone are insufficient to fully explain tumorigenesis. Emerging data suggest that DNA methylation might serve as a prognostic marker for invasive growth and recurrence, but further longitudinal studies are needed. This review highlights the need for future research to explore the methylome more thoroughly and to better define the clinical impact of epigenetic modifications in pituitary adenomas.
Collapse
Affiliation(s)
- Morten Winkler Møller
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Mathias Just Nortvig
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Mikkel Schou Andersen
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| |
Collapse
|
2
|
Dzialach L, Sobolewska J, Zak Z, Respondek W, Witek P. Prolactin-secreting pituitary adenomas: male-specific differences in pathogenesis, clinical presentation and treatment. Front Endocrinol (Lausanne) 2024; 15:1338345. [PMID: 38370355 PMCID: PMC10870150 DOI: 10.3389/fendo.2024.1338345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Prolactinomas (PRLomas) constitute approximately half of all pituitary adenomas and approximately one-fifth of them are diagnosed in males. The clinical presentation of PRLomas results from direct prolactin (PRL) action, duration and severity of hyperprolactinemia, and tumor mass effect. Male PRLomas, compared to females, tend to be larger and more invasive, are associated with higher PRL concentration at diagnosis, present higher proliferative potential, are more frequently resistant to standard pharmacotherapy, and thus may require multimodal approach, including surgical resection, radiotherapy, and alternative medical agents. Therefore, the management of PRLomas in men is challenging in many cases. Additionally, hyperprolactinemia is associated with a significant negative impact on men's health, including sexual function and fertility potential, bone health, cardiovascular and metabolic complications, leading to decreased quality of life. In this review, we highlight the differences in pathogenesis, clinical presentation and treatment of PRLomas concerning the male sex.
Collapse
Affiliation(s)
- Lukasz Dzialach
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Sobolewska
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Zak
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Wioleta Respondek
- Department of Internal Medicine, Endocrinology and Diabetes, Mazovian Brodnowski Hospital, Warsaw, Poland
| | - Przemysław Witek
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
Yearley AG, Chalif EJ, Gupta S, Chalif JI, Bernstock JD, Nawabi N, Arnaout O, Smith TR, Reardon DA, Laws ER. Metastatic pituitary tumors: an institutional case series. Pituitary 2023; 26:561-572. [PMID: 37523025 DOI: 10.1007/s11102-023-01341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE Pituitary carcinomas are a rare entity that respond poorly to multimodal therapy. Patients follow a variable disease course that remains ill-defined. METHODS We present an institutional case series of patients treated for pituitary carcinomas over a 30-year period from 1992 to 2022. A systematic review was conducted to identify prior case series of patients with pituitary carcinomas. RESULTS Fourteen patients with a mean age at pituitary carcinoma diagnosis of 52.5 years (standard deviation [SD] 19.4) met inclusion criteria. All 14 patients had tumor subtypes confirmed by immunohistochemistry and hormone testing, with the most common being ACTH-producing pituitary adenomas (n = 12). Patients had a median progression-free survival (PFS) of 1.4 years (range 0.7-10.0) and a median overall survival (OS) of 8.4 years (range 2.3-24.0) from pituitary adenoma diagnosis. Median PFS and OS were 0.6 years (range 0.0-2.2) and 1.5 years (range 0.1-9.6) respectively upon development of metastases. Most patients (n = 12) had locally invasive disease to the cavernous sinus, dorsum sellae dura, or sphenoid sinus prior to metastasis. Common sites of metastasis included the central nervous system, liver, lung, and bone. In a pooled analysis including additional cases from the literature, treatment of metastases with chemotherapy or a combination of radiation therapy and chemotherapy significantly prolonged PFS (p = 0.02), while failing to significantly improve OS (p = 0.14). CONCLUSION Pituitary carcinomas are highly recurrent, heterogenous tumors with variable responses to treatment. Multidisciplinary management with an experienced neuro-endocrine and neuro-oncology team is needed given the unrelenting nature of this disease.
Collapse
Affiliation(s)
- Alexander G Yearley
- Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| | - Eric J Chalif
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Saksham Gupta
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Joshua I Chalif
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Noah Nawabi
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Omar Arnaout
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Timothy R Smith
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - David A Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Edward R Laws
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Medina EJ, Zohdy YM, Porto E, Revuelta Barbero JM, Bray D, Maldonado J, Rodas A, Mayol M, Morales B, Neill S, Read W, Pradilla G, Ioachimescu A, Garzon-Muvdi T. Therapeutic response to pazopanib: case report and literature review on molecular abnormalities of aggressive prolactinomas. Front Endocrinol (Lausanne) 2023; 14:1195792. [PMID: 37529607 PMCID: PMC10388536 DOI: 10.3389/fendo.2023.1195792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction Aggressive prolactinomas (APRLs) pose a significant clinical challenge due to their high rate of regrowth and potentially life-threatening complications. In this study, we present a case of a patient with an APRL who had a trial of multiple therapeutic modalities with the aim to provide a review of molecular abnormalities and management of APRLs by corroborating our experience with previous literature. Methods A total of 268 articles were reviewed and 46 were included. Case reports and series, and studies that investigated the molecular and/or genetic analysis of APRLs were included. Special care was taken to include studies describing prolactinomas that would fall under the APRL subtype according to the European Society of Endocrinology guidelines; however, the author did not label the tumor as "aggressive" or "atypical". Addiontionally, we present a case report of a 56-year-old man presented with an invasive APRL that was resistant to multiple treatment modalities. Results Literature review revealed multiple molecular abnormalities of APRLs including mutations in and/or deregulation of ADAMTS6, MMP-9, PITX1, VEGF, POU6F2, CDKN2A, and Rb genes. Mismatch repair genes, downregulation of microRNAs, and hypermethylation of specific genes including RASSF1A, p27, and MGMT were found to be directly associated with the aggressiveness of prolactinomas. APRL receptor analysis showed that low levels of estrogen receptor (ER) and an increase in somatostatin receptors (SSTR5) and epidermal growth factor receptors (EGFR) were associated with increased invasiveness and higher proliferation activity. Our patient had positive immunohistochemistry staining for PD-L1, MSH2, and MSH6, while microarray analysis revealed mutations in the CDKN2A and POU6F2 genes. Despite undergoing two surgical resections, radiotherapy, and taking dopamine agonists, the tumor continued to progress. The patient was administered pazopanib, which resulted in a positive response and the patient remained progression-free for six months. However, subsequent observations revealed tumor progression. The patient was started on PD-L1 inhibitor pembrolizumab, yet the tumor continued to progress. Conclusion APRLs are complex tumors that require a multidisciplinary management approach. Knowledge of the molecular underpinnings of these tumors is critical for understanding their pathogenesis and identifying potential targets for precision medical therapy.
Collapse
Affiliation(s)
- Eduardo J. Medina
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Youssef M. Zohdy
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Edoardo Porto
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - David Bray
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Justin Maldonado
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Alejandra Rodas
- Department of Otolaryngology, Emory University, Atlanta, GA, United States
| | - Miguel Mayol
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Bryan Morales
- Department of Pathology, Emory University, Atlanta, GA, United States
| | - Stewart Neill
- Department of Pathology, Emory University, Atlanta, GA, United States
| | - William Read
- Department of Oncology, Emory University, Atlanta, GA, United States
| | - Gustavo Pradilla
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | | | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| |
Collapse
|
5
|
Targeting the Tumor Immune Microenvironment Could Become a Potential Therapeutic Modality for Aggressive Pituitary Adenoma. Brain Sci 2023; 13:brainsci13020164. [PMID: 36831707 PMCID: PMC9954754 DOI: 10.3390/brainsci13020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
OBJECT This study aimed to explore the relationship between the aggressiveness and immune cell infiltration in pituitary adenoma (PA) and to provide the basis for immuno-targeting therapies. METHODS One hundred and three patients with PA who underwent surgery at a single institution were retrospectively identified. The infiltration of macrophages and T-lymphocytes was quantitatively assessed. RESULTS The number of CD68+ macrophages was positively correlated with Knosp (p = 0.003) and MMP-9 expression grades (p = 0.00). The infiltration of CD163+ macrophages differed among Knosp (p = 0.022) and MMP-9 grades (p = 0.04). CD8+ tumor-infiltrating lymphocytes (TILs) were also positively associated with Knosp (p = 0.002) and MMP-9 grades (p = 0.01). Interestingly, MGMT expression was positively correlated with MMP-9 staining extent (p = 0.000). The quantities of CD8+ TILs (p = 0.016), CD68+ macrophages (p = 0.000), and CD163+ macrophages (p = 0.043) were negatively associated with MGMT expression levels. The number of CD68+ macrophages in the PD-L1 negative group was significantly more than that in the PD-L1 positive group (p = 0.01). The rate of PD-L1 positivity was positively correlated with the Ki-67 index (p = 0.046) and p53 expression (p = 0.029). CONCLUSION Targeted therapy for macrophages and CD8+ TILs could be a helpful treatment in the future for aggressive PA. Anti-PD-L1 therapy may better respond to PAs with higher Ki-67 and p53 expression and more infiltrating CD68+ macrophages. Multiple treatment modalities, especially combined with immunotherapy could become a novel therapeutic strategy for aggressive PA.
Collapse
|
6
|
Kontogeorgos G, Thodou E, Osamura RY, Lloyd RV. High-risk pituitary adenomas and strategies for predicting response to treatment. Hormones (Athens) 2022; 21:1-14. [PMID: 35061210 DOI: 10.1007/s42000-021-00333-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/18/2021] [Indexed: 12/30/2022]
Abstract
High-risk pituitary adenomas are aggressive. They show clinical and imaging features similar to those of carcinomas, including infiltration of the surrounding brain structures, but lack cerebrospinal or systemic metastases. In addition, they display distinct behavior, including tendency for fast growth and frequent recurrences, which are difficult to control. The term "high-risk" adenoma was first introduced in the 4th edition of the World Health Organization Classification of Endocrine Tumors in 2017. Five defined adenoma types belong to this category, including sparsely granulated somatotroph, lactotroph in men, Crooke cell, silent corticotroph, and plurihormonal PIT-1 positive adenomas. The morphological and immunohistochemical characteristics of high-risk adenomas are herein described in detail. In addition, the clinical features and the treatment options are presented. This review focuses on predictive markers assessed by immunohistochemistry, which help clinicians to design the appropriate treatment strategies for high-risk adenomas. Somatostatin receptor status predicts effectiveness of postsurgical treatment with somatostatin analogs, and MGMT expression predicts response to treatment with temozolomide. This comprehensive review presents the clinical and pathological features of high-risk pituitary adenomas, underlines the contribution of immunohistochemistry, and emphasizes the leading role of pathology in the design of optimal clinical management.
Collapse
Affiliation(s)
- George Kontogeorgos
- Division of Endocrinology, First Propaedeutic Department of Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens, Athens, Greece.
- Department of Pathology and Pituitary Tumor Reference Center, G. Gennimatas General Hospital of Athens, Athens, Greece.
| | - Eleni Thodou
- Department of Pathology, University of Thessaly, Larissa, Greece
| | - Robert Y Osamura
- Department of Pathology, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Ricardo V Lloyd
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
7
|
Saeger W, Koch A. Clinical Implications of the New WHO Classification 2017 for Pituitary Tumors. Exp Clin Endocrinol Diabetes 2021; 129:146-156. [PMID: 33690870 DOI: 10.1055/a-1310-7900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
According to the WHO classification 2017 of Pituitary Tumors adenomas are classified not only by structure and immunostaining for pituitary hormones but also by expression of the pituitary transcription factors Pit-1, T-pit and SF-1. By these factors, three cell lineages can be identified: Pit-1 for the GH-, Prolactin- and TSH-cell lineage, T-pit for the ACTH-cell lineage, and SF-1 for the gonadotrophic cell lineage. By this principle, all GH and/or Prolactin producing and all TSH producing adenomas must be positive for Pit-1, all corticotrophic adenomas for T-pit, and all gonadotrophic for SF-1. In adenomas without expression of pituitary hormones immunostainings for the transcription factors have to be examined. If these are also negative the criteria for an endocrine inactive null cell adenoma are fulfilled. If one transcription factor is positive the corresponding cell lineage indicates a potential hormonal activity of the adenoma. So Pit-1 expressing hormone-negative adenomas can account for acromegaly, hyperprolactinemia, or TSH hyperfunction. T-pit positive hormone negative adenomas can induce Cushing's disease, and SF-1 positive hormone negative tumors indicate gonadotrophic adenomas. Instead of the deleted atypical adenoma of the WHO classification of 2004 now (WHO classification 2017) criteria exist for the identification of aggressive adenomas with a conceivably worse prognosis. Some adenoma subtypes are described as aggressive "per se" without necessity of increased morphological signs of proliferation. All other adenoma subtypes must also be designated as aggressive if they show signs of increased proliferation (mitoses, Ki-67 index>3-5%, clinically rapid tumor growth) and invasion. By these criteria about one third of pituitary adenoma belong to the group of aggressive adenomas with potentially worse prognosis. The very rare pituitary carcinoma (0.1 % of pituitary tumors) is defined only by metastases. Many of them develop after several recurrences of Prolactin or ACTH secreting adenomas. The correlation of clinical findings and histological classification of pituitary adenomas is very important since every discrepancy has to be discussed between clinicians and pathologists. Based on data of the German Registry of Pituitary Tumors a table for examinations of correlations is shown in this review.
Collapse
Affiliation(s)
- Wolfgang Saeger
- Institute of Pathology and Neuropathology of the University of Hamburg, UKE, Hamburg, Germany
| | - Arend Koch
- Institute of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
8
|
Das L, Gupta N, Dutta P, Walia R, Vaiphei K, Rai A, Radotra BD, Gupta K, Sreedharanunni S, Ahuja CK, Bhansali A, Tripathi M, Sood R, Dhandapani S. Early Initiation of Temozolomide Therapy May Improve Response in Aggressive Pituitary Adenomas. Front Endocrinol (Lausanne) 2021; 12:774686. [PMID: 34975752 PMCID: PMC8718901 DOI: 10.3389/fendo.2021.774686] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/08/2021] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Aggressive pituitary adenomas (APAs) are, by definition, resistant to optimal multimodality therapy. The challenge lies in their early recognition and timely management. Temozolomide is increasingly being used in patients with APAs, but evidence supporting a favorable response with early initiation is lacking. METHODS This was a single-center study of all patients with APAs who received at least 3 cycles of temozolomide (150-200 mg/m2). Their baseline clinico-biochemical and radiological profiles were recorded. Immunohistochemical evaluation for cell-cycle markers O6-methylguanine-DNA methyltransferase (MGMT), MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), MutL homolog 1 (MLH1), and postmeiotic segregation increased 2 (PMS2) was performed, and h-scores (product of the number of positive cells and staining intensity) were calculated. Response was assessed in terms of radiological response using the RECIST criteria. Patients with controlled disease (≥30% reduction in tumor volume) were classified as responders. RESULTS The study comprised 35 patients (48.6% acromegaly, 37.1% prolactinomas, and 14.3% non-functioning pituitary adenomas). The median number of temozolomide (TMZ) cycles was 9 (IQR 6-14). Responders constituted 68.6% of the cohort and were more likely to have functional tumors, a lower percentage of MGMT-positive staining cells, and lower MGMT h-scores. There was a significantly longer lag period in the initiation of TMZ therapy in non-responders as compared with responders (median 36 vs. 15 months, p = 0.01). ROC-derived cutoffs of 31 months for the duration between diagnosis and TMZ initiation, low-to-intermediate MGMT positivity (40% tumor cells), and MGMT h-score of 80 all had a sensitivity exceeding 80% and a specificity exceeding 70% to predict response. CONCLUSION Early initiation of TMZ therapy, functional tumors, and low MGMT h-score predict a favorable response to TMZ in APAs.
Collapse
Affiliation(s)
- Liza Das
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Nidhi Gupta
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Pinaki Dutta
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
- *Correspondence: Pinaki Dutta,
| | - Rama Walia
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Kim Vaiphei
- Department of Histopathology, PGIMER, Chandigarh, India
| | - Ashutosh Rai
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Kirti Gupta
- Department of Histopathology, PGIMER, Chandigarh, India
| | | | | | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Ridhi Sood
- Department of Histopathology, PGIMER, Chandigarh, India
| | | |
Collapse
|
9
|
Nakano-Tateno T, Lau KJ, Wang J, McMahon C, Kawakami Y, Tateno T, Araki T. Multimodal Non-Surgical Treatments of Aggressive Pituitary Tumors. Front Endocrinol (Lausanne) 2021; 12:624686. [PMID: 33841328 PMCID: PMC8033019 DOI: 10.3389/fendo.2021.624686] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Up to 35% of aggressive pituitary tumors recur and significantly affect mortality and quality of life. Management can be challenging and often requires multimodal treatment. Current treatment options, including surgery, conventional medical therapies such as dopamine agonists, somatostatin receptor agonists and radiotherapy, often fail to inhibit pituitary tumor growth. Recently, anti-tumor effects of chemotherapeutic drugs such as Temozolomide, Capecitabine, and Everolimus, as well as peptide receptor radionuclide therapy on aggressive pituitary tumors have been increasingly investigated and yield mixed, although sometimes promising, outcomes. The purpose of this review is to provide thorough information on non-surgical medical therapies and their efficacies and used protocols for aggressive pituitary adenomas from pre-clinical level to clinical use.
Collapse
Affiliation(s)
- Tae Nakano-Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kheng Joe Lau
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Justin Wang
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, United States
| | - Cailin McMahon
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, United States
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Toru Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Toru Tateno, ; Takako Araki,
| | - Takako Araki
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Toru Tateno, ; Takako Araki,
| |
Collapse
|
10
|
Kobyakov GL, Chernov IV, Astafieva LI, Trunin YY, Poddubsky AA, Kalinin PL. [Use of chemotherapy in the treatment of aggresive pituitary adenomas]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2020; 84:69-75. [PMID: 32207745 DOI: 10.17116/neiro20208401169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIM To clarify the concept of 'aggressive pituitary adenoma' using analysis of the current concepts, as well as to determine the optimal treatment algorithm for this disease and the place of chemotherapy in this treatment. Pituitary adenomas comprise from 10 to 15% of intracranial neoplasms. Despite the fact that pituitary adenomas are benign neoplasms, in 25-55% of cases they demonstrate invasive growth, growing into the surrounding structures (sphenoid sinus, cavernous sinus, etc.). Due to the lack of a standard definition of aggressive pituitary adenomas (due to the lack of clear criteria for this disease), there are no studies in the literature reporting optimal treatment for this group of patients, except for several publications describing the use of Temozolomide as palliative therapy.
Collapse
Affiliation(s)
| | - I V Chernov
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | | | | | - P L Kalinin
- Burdenko Neurosurgical Center, Moscow, Russia
| |
Collapse
|
11
|
Burman P, Lamb L, McCormack A. Temozolomide therapy for aggressive pituitary tumours - current understanding and future perspectives. Rev Endocr Metab Disord 2020; 21:263-276. [PMID: 32147777 DOI: 10.1007/s11154-020-09551-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of temozolomide (TMZ) for the management of aggressive pituitary tumours (APT) has revolutionised clinical practice in this field with significantly improved clinical outcomes and long-term survival. Its use is now well established however a large number of patients do not respond to treatment and recurrence after cessation of TMZ is common. A number of challenges remain for clinicians such as appropriate patient selection, treatment duration and the role of combination therapy. This review will examine the use of TMZ to treat APT including mechanism of action, treatment regimen and duration; biomarkers predicting response to treatment and patient selection; and current evidence for administration of TMZ in combination with other agents.
Collapse
Affiliation(s)
- Pia Burman
- Department of Endocrinology, Skåne University Hospital, University of Lund, Malmö, Sweden
| | - Lydia Lamb
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Ann McCormack
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia.
- Garvan Institute of Medical Research, Sydney, NSW, Australia.
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
12
|
Elbelt U, Schlaffer SM, Buchfelder M, Knappe UJ, Vila G, Micko A, Deutschbein T, Unger N, Lammert A, Topuzoglu-Müller T, Bojunga J, Droste M, Johanssen S, Kolenda H, Ritzel K, Buslei R, Strasburger CJ, Petersenn S, Honegger J. Efficacy of Temozolomide Therapy in Patients With Aggressive Pituitary Adenomas and Carcinomas-A German Survey. J Clin Endocrinol Metab 2020; 105:5634134. [PMID: 31746334 DOI: 10.1210/clinem/dgz211] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/18/2019] [Indexed: 11/19/2022]
Abstract
CONTEXT Despite growing evidence that temozolomide (TMZ) therapy is effective for the treatment of aggressive pituitary tumors (APTs) or carcinomas (PCs), individual therapy decisions remain challenging. OBJECTIVE We therefore aimed to report on clinical characteristics leading to initiation of TMZ therapy and to add evidence on TMZ long-term effectiveness. DESIGN AND SUBJECTS Retrospective survey on TMZ treatment in patients with APTs or PCs. TMZ therapy was initiated in 47 patients (22 females) with APTs (n = 34) or PCs (n = 13). Mean age at diagnosis was 45 ± 15 years. The immunohistochemical subtypes were corticotroph (n = 20), lactotroph (n = 18), and nonfunctioning (n = 9) tumors. TMZ therapy started 8 years after initial diagnosis using a standard regimen (median 6 cycles) for the majority of patients. RESULTS Long-term radiological response to TMZ after a median follow-up of 32 months with 4 patients still on TMZ therapy was tumor regression for 9 (20%), stable disease for 8 (17%), and tumor progression for 29 patients (63%) (outcome data available for 46 patients). Progression occurred 16 months after initiation of TMZ. Median estimated progression-free survival was 23 months. Disease stabilization and median progression-free survival did not differ between patients with APTs or PCs. Predictors of tumor response were not identified. Overall, TMZ was well tolerated. CONCLUSION We performed a nationwide survey on TMZ therapy in patients with APTs and PCs. While early response rates to TMZ are promising, long-term outcome is less favorable. Prolonged TMZ administration should be considered. We were not able to confirm previously reported predictors of tumor response to TMZ.
Collapse
Affiliation(s)
- Ulf Elbelt
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sven M Schlaffer
- Department of Neurosurgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ulrich J Knappe
- Department of Neurosurgery, Johannes Wesling Klinikum, Universitätsklinikum der Ruhruniversität Bochum, Minden, Germany
| | - Greisa Vila
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University Vienna, Vienna, Austria
| | - Alexander Micko
- Department of Neurosurgery, Medical University Vienna, Vienna, Austria
| | - Timo Deutschbein
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, University of Würzburg, Würzburg, Germany
| | - Nicole Unger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, Essen, Germany
| | - Alexander Lammert
- Vth Department of Medicine, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Tengü Topuzoglu-Müller
- Department of Endocrinology, Diabetes and Preventive Medicine, University Hospital of Cologne, Cologne, Germany
| | - Jörg Bojunga
- Department of Internal Medicine 1, Johann Wolfgang Goethe-University Hospital, Frankfurt, Germany
| | | | | | - Herbert Kolenda
- Department of Neurosurgery, Agaplesion Diakonieklinikum Rotenburg, Rotenburg, Germany
| | - Katrin Ritzel
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rolf Buslei
- Institute of Pathology, SozialStiftung Bamberg, Bamberg, Germany
| | - Christian J Strasburger
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Jürgen Honegger
- Department of Neurosurgery, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
13
|
Kontogeorgos G, Thodou E, Koutourousiou M, Kaltsas G, Seretis A. MGMT immunohistochemistry in pituitary tumors: controversies with clinical implications. Pituitary 2019; 22:614-619. [PMID: 31571099 DOI: 10.1007/s11102-019-00993-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Temozolomide (TMZ) is currently considered as a rational therapeutic option for patients with progressively aggressive pituitary adenomas and carcinomas not responding to conventional therapies. Administration of TMZ results in clinical response and improvement in survival of many of these patients depending upon the expression of the DNA repair enzyme O-6 methylguanine DNA transferase (MGMT). Low or negative MGMT immunoreactivity predicts responsiveness to TMZ therapy. Therefore, MGMT serves as a criterion to select candidate patients anticipating response to treatment. MATERIALS AND METHODS The MGMT expression was investigated in 25 pituitary adenomas with Ki-67 labeling index more that 3% and p53 expression, using various antigen retrieval protocols. After direct application of the antibody, only one adenoma yielded positive for MGMT. However, after pretreatment of tissue sections with antigen retrieval protocols, another 3 adenomas, initially negative turned to positive. CONCLUSIONS These findings could explain lack of response to TMZ treatment in patients with false negative MGMT immunohistochemistry. Evaluation of tumor samples for MGMT expression should carefully be carried-out using the optimum immunohistochemical protocol to obtain consistent and reliable results that help to identify patients that could respond to TMZ therapy.
Collapse
Affiliation(s)
- George Kontogeorgos
- First Propaepeudic Department of Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Attica, Greece.
- Department of Pathology, "G. Gennimatas" Athens General Hospital, 154 Messogion Ave., 11527, Athens, Attica, Greece.
| | - Eleni Thodou
- Department of Pathology, School of Medicine, University of Thessaly, Viopolis, 41110, Larissa, Thessaly, Greece
| | - Mary Koutourousiou
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY, 40202, USA
| | - Gregory Kaltsas
- First Propaepeudic Department of Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Attica, Greece
| | - Andreas Seretis
- Neurosurgery Department, Henry Dunant Hospital, 107 Messogion Ave., 11527, Athens, Attica, Greece
- Department of Neurosurgery, "G. Gennimatas" Athens General Hospital, 154 Messogion Ave., 11527, Athens, Attica, Greece
| |
Collapse
|
14
|
Kontogeorgos G, Thodou E. Is MGMT the best marker to predict response of temozolomide in aggressive pituitary tumors? Alternative markers and prospective treatment modalities. Hormones (Athens) 2019; 18:333-337. [PMID: 31721137 DOI: 10.1007/s42000-019-00145-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/15/2019] [Indexed: 01/02/2023]
Affiliation(s)
- George Kontogeorgos
- First Propaedeutic Department of Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens, 75 Mikras Asias Str., Athens, 11527, Attica, Greece.
- Department of Pathology, "G. Gennimatas" Athens General Hospital, Athens, Greece.
| | - Eleni Thodou
- Department of Pathology, University of Thessaly, School of Medicine, Larissa, Greece
| |
Collapse
|
15
|
Micko ASG, Höftberger R, Wöhrer A, Millesi M, Knosp E, Wolfsberger S. MGMT assessment in pituitary adenomas: comparison of different immunohistochemistry fixation chemicals. Pituitary 2018; 21:266-273. [PMID: 29344904 PMCID: PMC5942339 DOI: 10.1007/s11102-018-0862-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Despite the established role of O6-methyl-guanine-DNA methyltransferase (MGMT) as a marker for temozolomide response, consensus of the most reliable method to assess MGMT expression in pituitary adenomas is still missing. Currently, immunohistochemistry (IHC) assessment of formaldehyde fixed tissue samples is most widely used in a semiquantitative description. As formaldehyde fails to completely preserve nucleic acids, RCL2, an alcohol-based formaldehyde-free fixative, has been proposed as a more reliable alternative in terms of cell stability. Furthermore, as the current method of IHC is semiquantitative and observer-dependent, pyrosequencing, an objective tool to evaluate the methylation status of the MGMT promoter, has emerged as a reliable and accurate alternative. The aim of this study was to validate the current IHC method for assessment of MGMT protein expression in pituitary adenomas. METHODS The tissue samples of 8 macroadenomas with positive IHC MGMT expression (> 50%) were investigated: first, we compared the time dependent stability of MGMT protein expression after pituitary adenoma removal between formaldehyde vs. RCL2. Then, we compared positive IHC MGMT expression with methylated promoter status using pyrosequencing. RESULTS In the first 12 h after adenoma removal, tissue samples remained MGMT positive in significantly more samples when fixated with formaldehyde than with RCL2, respectively (96 vs. 81%, p = 0.025). CONCLUSION Our data confirm that the current method using formaldehyde tissue fixation and IHC reveals stable and reliable results of MGMT assessment in pituitary adenomas.
Collapse
Affiliation(s)
- Alexander S G Micko
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Adelheid Wöhrer
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Matthias Millesi
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria
| | - Engelbert Knosp
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria.
| | - Stefan Wolfsberger
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria
| |
Collapse
|
16
|
Micko ASG, Wöhrer A, Höftberger R, Vila G, Marosi C, Knosp E, Wolfsberger S. MGMT and MSH6 immunoexpression for functioning pituitary macroadenomas. Pituitary 2017; 20:643-653. [PMID: 28900805 PMCID: PMC5655586 DOI: 10.1007/s11102-017-0829-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE Knowledge of biological behavior is crucial for clinical management of functioning pituitary macroadenomas. For recurrent cases unresponsive to standard treatment, temozolomide (TMZ) has been used as a therapeutic alternative. MGMT (O6-methyl-guanine-DNA methyltransferase) and MSH6 (mutS homolog 6) immunoexpression have been linked to the response to TMZ treatment and MGMT immunoexpression has been additionally linked to early recurrence of non-functioning pituitary adenomas. The aim of this study was to assess the prognostic value of MGMT and MSH6 immunoexpression for aggressive functioning pituitary adenomas. METHODS The study cohort comprised a single center series of 76 patients who underwent an operation for functioning pituitary macroadenoma. We retrospectively compared 38 patients with postoperative persistent or recurrent disease with another set of 38 patients who were in endocrine remission. RESULTS Low-to-moderate MGMT immunoexpression (<50%) was significantly more frequent in the group with persistent/recurrent disease than in cases of endocrine remission (66 vs. 21%, p < 0.001). Furthermore, adenomas with low-to-moderate MGMT immunoexpression were significantly more often recurrent (76 vs. 30%, p < 0.001) and invasive (64 vs. 28%, p = 0.002). CONCLUSION In our series, low-to-moderate MGMT immunoexpression was the only marker that significantly correlated with surgical invasiveness and recurrence in functioning pituitary macroadenomas. Therefore, in the future, MGMT status may be considered an additional marker for understanding the biological behavior of pituitary adenomas.
Collapse
Affiliation(s)
- Alexander S G Micko
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria
| | - Adelheid Wöhrer
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | | | - Greisa Vila
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Christine Marosi
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Engelbert Knosp
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria.
| | - Stefan Wolfsberger
- Department of Neurosurgery, Medical University Vienna, Waehringer Guertel 18-20, 1097, Vienna, Austria
| |
Collapse
|
17
|
O-6-Methylguanine-DNA methyltransferase expression is associated with pituitary adenoma tumor recurrence: a systematic meta-analysis. Oncotarget 2017; 8:19674-19683. [PMID: 28152515 PMCID: PMC5386713 DOI: 10.18632/oncotarget.14936] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022] Open
Abstract
O-6-methylguanine-DNA methyltransferase (MGMT) reportedly counteracts the cytotoxic effects of the alkylating agent temozolomide. MGMT expression is often low in aggressive pituitary adenomas (PAs) and recurrent PAs. However, because these associations are controversial, we performed this meta-analysis to clarify the involvement of MGMT in the prognosis and clinicopathology of PA. We searched for relevant studies in electronic databases (MEDLINE, the Cochrane Library Database, EMBASE, CINAHL, Web of Science and the Chinese Biomedical Database (CBD)) and calculated/pooled the odds ratios (ORs) or standard mean differences (SMDs) with 95% confidence intervals (95% CIs). Eleven case-control studies with a total of 454 PA patients were included. Our meta-analysis revealed that lower expression of MGMT was associated with PA recurrence (OR=2.09, 95% CI=1.09-4.02; p=0.026). On the other hand, MGMT expression was not associated with PA invasiveness (OR=1.112, 95% CI=0.706-1.753; p=0.646), Unexpectedly, MGMT expression could not be used to distinguish functional from non-functional PA patients (OR=1.766, 95% CI=0.938-3.324; p=0.078). The MGMT expression was not found to be related to other clinicopathological indicators of PA including age, gender or tumor size. No publication bias was detected in this meta-analysis (p>0.05). This meta-analysis suggests that MGMT expression may be associated with PA tumor recurrence, but not be related to invasiveness or other clinicopathological indicators. Thus, detection of MGMT expression may facilitate outcome prediction and guide clinical therapy for PA patients.
Collapse
|
18
|
Angelousi A, Dimitriadis GK, Zografos G, Nölting S, Kaltsas G, Grossman A. Molecular targeted therapies in adrenal, pituitary and parathyroid malignancies. Endocr Relat Cancer 2017; 24:R239-R259. [PMID: 28400402 DOI: 10.1530/erc-16-0542] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022]
Abstract
Tumourigenesis is a relatively common event in endocrine tissues. Currently, specific guidelines have been developed for common malignant endocrine tumours, which also incorporate advances in molecular targeted therapies (MTT), as in thyroid cancer and in gastrointestinal neuroendocrine malignancies. However, there is little information regarding the role and efficacy of MTT in the relatively rare malignant endocrine tumours mainly involving the adrenal medulla, adrenal cortex, pituitary, and parathyroid glands. Due to the rarity of these tumours and the lack of prospective studies, current guidelines are mostly based on retrospective data derived from surgical, locoregional and ablative therapies, and studies with systemic chemotherapy. In addition, in many of these malignancies the prognosis remains poor with individual patients responding differently to currently available treatments, necessitating the development of new personalised therapeutic strategies. Recently, major advances in the molecular understanding of endocrine tumours based on genomic, epigenomic, and transcriptome analysis have emerged, resulting in new insights into their pathogenesis and molecular pathology. This in turn has led to the use of novel MTTs in increasing numbers of patients. In this review, we aim to present currently existing and evolving data using MTT in the treatment of adrenal, pituitary and malignant parathyroid tumours, and explore the current utility and effectiveness of such therapies and their future evolution.
Collapse
Affiliation(s)
- Anna Angelousi
- Department of PathophysiologySector of Endocrinology, National & Kapodistrian University of Athens, Athens, Greece
| | - Georgios K Dimitriadis
- Division of Translational and Experimental MedicineUniversity of Warwick Medical School, Clinical Sciences Research Laboratories, Coventry, UK
| | - Georgios Zografos
- Third Department of SurgeryAthens General Hospital "Georgios Gennimatas", Athens, Greece
| | - Svenja Nölting
- Department of Internal Medicine IICampus Grosshadern, University-Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Gregory Kaltsas
- Department of PathophysiologySector of Endocrinology, National & Kapodistrian University of Athens, Athens, Greece
- Division of Translational and Experimental MedicineUniversity of Warwick Medical School, Clinical Sciences Research Laboratories, Coventry, UK
- Department of EndocrinologyOxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | - Ashley Grossman
- Department of EndocrinologyOxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Jordan JT, Miller JJ, Cushing T, Seijo M, Batchelor TT, Arrillaga-Romany IC, Shih HA, Nachtigall LB, Loeffler JS, Dietrich J. Temozolomide therapy for aggressive functioning pituitary adenomas refractory to surgery and radiation: a case series. Neurooncol Pract 2017; 5:64-68. [PMID: 31385986 DOI: 10.1093/nop/npx013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Treatment of aggressive pituitary adenomas typically involves a multimodality approach based on histopathological features and may include pharmacotherapy, surgery, and occasionally radiation therapy. In cases of treatment-refractory tumor progression, chemotherapy may be considered; however, no standard chemotherapeutic regimen has been established. Literature review suggests that temozolomide may have a beneficial role in a subset of cases. To understand the efficacy of temozolomide in progressive pituitary tumors, we reviewed the outcomes of cases at our center. Methods We performed a retrospective chart review to report the outcome and unique features of 7 patients with aggressive functioning pituitary adenomas or carcinomas treated with temozolomide. Tumor pathology included somatotroph (n = 1), corticotroph (n = 3), and lactotroph (n = 3) tumors. Results Four of the 7 patients had at least 2 prior resections, and all had prior radiation and surgery before treatment with temozolomide. Notably, all patients showed response to therapy, defined as either stable disease (43%) or partial response (57%). Median progression-free survival was 1.66 years, and median overall survival was 4 years. Conclusion Our data suggest that temozolomide has an important role in the management of aggressive functioning pituitary tumors that are resistant to standard therapies, and that optimization of therapy with temozolomide may involve individualized regimens. Future prospective clinical trials should be considered.
Collapse
Affiliation(s)
- Justin T Jordan
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| | - Julie J Miller
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| | - Tucker Cushing
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| | - Marlon Seijo
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| | - Tracy T Batchelor
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| | | | - Helen A Shih
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA.,Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Lisa B Nachtigall
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA.,Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA
| | - Jay S Loeffler
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA.,Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Jorg Dietrich
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
20
|
Abstract
PURPOSE Uncontrolled acromegaly results in approximately 2-fold excess mortality. Pituitary surgery is first-line therapy, and medical treatment is indicated for persistent disease. While cabergoline and pegvisomant are used in select patients, somatostatin receptor ligands (SRLs) remain the cornerstone of medical treatment. Management of patients poorly responsive to SRLs is therefore, challenging. The purpose of this review is to highlight the options for combination medical therapy in the treatment of acromegaly, with an emphasis on efficacy and safety. METHODS All original articles/abstracts detailing combination medical therapy in acromegaly were identified from a PubMed search. RESULTS Studies reviewed included retrospective and open-label prospective studies. While the combination of SRL and cabergoline was generally well tolerated, a lower baseline insulin-like growth factor-1 (IGF-1) level was the best predictor of efficacy; this combination may be most effective in patients with mildly elevated IGF-1. SRL-pegvisomant combination normalized IGF-1 in the majority of patients; continued efficacy despite individual drug dosing reduction was also reported. The risk of significant liver enzyme elevation was, however, higher than that reported with SRL monotherapy; close monitoring is recommended. Data on pegvisomant-cabergoline combination is limited, but this may be an option in the setting of SRL intolerance. Reports on temozolomide used in combination with other medical therapies in patients with aggressive GH-secreting tumors are also summarized. CONCLUSION While more prospective, randomized controlled trials on long-term efficacy and safety are needed, combination medical therapy remains a treatment strategy that should be considered for acromegaly patients poorly responsive to SRLs.
Collapse
Affiliation(s)
- Dawn Shao Ting Lim
- Departments of Medicine (Endocrinology) and Neurological Surgery, and Northwest Pituitary Center, Oregon Health & Science University, 3303 SW Bond Ave, Mail Code CH8N, Portland, OR, 97239, USA
| | - Maria Fleseriu
- Departments of Medicine (Endocrinology) and Neurological Surgery, and Northwest Pituitary Center, Oregon Health & Science University, 3303 SW Bond Ave, Mail Code CH8N, Portland, OR, 97239, USA.
| |
Collapse
|
21
|
Basaran R, Onoz M, Bolukbasi FH, Efendioglu M, Sav A. LOW O6-METHYLGUANINE-DNA METHYTRANSFERASE (MGMT) AND PAN-CYTOKERATIN (PAN-CK) EXPRESSION VIA IMMUNOHISTOCHEMISTRY IN PITUITARY ADENOMAS. ACTA ENDOCRINOLOGICA-BUCHAREST 2017; 13:282-293. [PMID: 31149189 DOI: 10.4183/aeb.2017.282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Introduction Pituitary adenomas (PA) are the third most common intracranial tumors, with an incidence rate of 10-15%. More than half are invasive, infiltrating adjacent structures. The primary objective of this project was to determine whether MGMT expression is associated with the invasiveness of PA. Material and Method All patients who underwent surgical decompression consecutively between 2007-2012 were included. All data were obtained from the case records. Formalin-fixed paraffin-embedded (FFPE) tissue specimens were stained with hematoxylin and eosin (HE) and then examined via light microscope. Paraffin blocks that lacked necrosis and hemorrhage were chosen for histologic examination. In addition to an immunoprofile battery that consisted of Ki-67 and p53, MGMT, S-100 and Pan-CK were evaluated as well. Results The subjects included 25 women and 15 men. The mean age was 48.9 ± 14.5 years. Of these, 63% of cases involved the invasion of adjacent structures. Of the PA, 17 (42%) were non-functioning pituitary adenomas (NFPA). There was a statistically significant relationship between the invasiveness and Ki-67, p53, MGMT expression, and prolactinoma. Gonodotropinomas were mostly non-invasive. FPAs presented invasive features more frequently than NFPAs. Pan-CK was positive in GH-secreting adenomas but negative in FSH- and LH-secreting adenomas. Conclusion Ki-67 and p53 in lower expression level can be used for evaluating invasiveness but not for recurrence. MGMT expression can be a useful IHC indicator for invasiveness. However, Pan-CK cannot be used for invasiveness or aggressiveness.
Collapse
Affiliation(s)
- R Basaran
- Istanbul Medeniyet University Goztepe Education and Research Hospital, Department of Neurosurgery, Istanbul, Turkey
| | - M Onoz
- Medipol University School of Medicine, Department of Neurosurgery, Istanbul, Turkey
| | - F H Bolukbasi
- Medipol University School of Medicine, Department of Neurosurgery, Istanbul, Turkey
| | - M Efendioglu
- Istanbul Medeniyet University Goztepe Education and Research Hospital, Department of Neurosurgery, Istanbul, Turkey
| | - A Sav
- Acibadem University School of Medicine, Department of Pathology, Istanbul, Turkey
| |
Collapse
|
22
|
Aggressive Pituitary Adenomas: The Dark Side of the Moon. World Neurosurg 2017; 97:140-155. [DOI: 10.1016/j.wneu.2016.09.092] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 01/14/2023]
|
23
|
Zheng X, Li S, Zhang W, Zang Z, Hu J, Yang H. Current biomarkers of invasive sporadic pituitary adenomas. ANNALES D'ENDOCRINOLOGIE 2016; 77:658-667. [PMID: 27659267 DOI: 10.1016/j.ando.2016.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/29/2016] [Accepted: 02/21/2016] [Indexed: 12/22/2022]
Abstract
Though pituitary adenomas (PA) are considered benign, some of them exhibit invasive behaviors such as recurrence and low rate of total surgical resection. Reliable prognostic biomarkers for invasive PA are highly desired; however they remain to be identified. In this review, we summarize the current controversial findings of biomarkers for invasive sporadic PA, and we discuss the possible reasons for the controversies.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Weihua Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Zhenle Zang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Jintao Hu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China.
| |
Collapse
|
24
|
Grandidge C, Hall A, Kitchen N. Secreting Follicle-Stimulating Hormone Pituitary Carcinoma with Cervical Metastasis. World Neurosurg 2016; 93:490.e13-6. [PMID: 27449685 DOI: 10.1016/j.wneu.2016.05.095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Pituitary carcinoma is defined by either metastases outside the central nervous system or noncontiguous foci within the central nervous system. This case report details the first documented case of a pathologically isolated follicle-stimulating hormone-secreting pituitary carcinoma and its presentation of metastasis. CASE DESCRIPTION A 63-year-old man developed intrascapular pain radiating up his neck to his occiput. He had undergone a transsphenoidal hypophysectomy 2 years previously for an atypical pituitary macroadenoma. Subsequent magnetic resonance imaging identified a focal, solitary, well-circumscribed, homogeneous T2 high-signal intradural, extramedullary enhancing mass at C2-3 in a right ventral parasagittal location, extending toward the exit foramina. On surgical excision with a laminectomy, the mass demonstrated a morphologic appearance of a malignant neuroendocrine tumor with an immunoprofile similar to the original atypical pituitary adenoma. This was in keeping with metastatic disease secondary to a follicle-stimulating hormone-secreting pituitary carcinoma. CONCLUSIONS Although rare, metastatic spread is recognized in patients with atypical pituitary adenoma. This should form the differential diagnosis for such patients presenting with symptoms that could be attributed to metastatic lesions within the neuraxis. In these patients, who undergo regular surveillance in joint neuroendocrine clinics, more urgent investigation of new spinal pain should be instigated to exclude metastatic disease.
Collapse
Affiliation(s)
- Carly Grandidge
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.
| | - Andy Hall
- Royal National Throat, Nose and Ear Hospital, London, United Kingdom
| | - Neil Kitchen
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology & Neurosurgery, London, United Kingdom
| |
Collapse
|
25
|
Moisi M, Cruz AS, Benkers T, Rostad S, Broyles FB, Yuen K, Mayberg M. Treatment of Aggressive Prolactin-Secreting Pituitary Adenomas with Adjuvant Temozolomide Chemotherapy: A Review. Cureus 2016; 8:e658. [PMID: 27489751 PMCID: PMC4963231 DOI: 10.7759/cureus.658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Most prolactin-secreting pituitary adenomas demonstrate slow growth and are effectively managed with medical/surgical therapy. Rarely, these tumors can behave aggressively with rapid growth and invasion of local tissues, and are refractory to medical, surgical, or radio-surgical therapies. We report a case of a prolactin-secreting adenoma in a young woman, which became progressively aggressive and refractory to usual treatment modalities, but responded to treatment with the chemotherapeutic agent temozolomide. In addition, we review the literature for treatment of refractory adenomas with temozolomide. The clinical and pathologic characteristics of aggressive prolactin-secreting adenomas are reviewed, as well as their response to dopamine agonists, surgery, radiotherapy, and chemotherapy.
Collapse
Affiliation(s)
- Marc Moisi
- Seattle Science Foundation ; Neurological Surgery, Wayne State University
| | - Aurora S Cruz
- Neurological Surgery, University of Louisville ; Neurological Surgery, University of California, Irvine ; Neurological Surgery, Swedish Neuroscience Institute
| | | | | | | | - Kevin Yuen
- Swedish Pituitary Center, Swedish Neuroscience Institute
| | | |
Collapse
|
26
|
Lasolle H, Raverot G. Therapeutic innovations in endocrine diseases – part 3 : temozolomide and future therapeutics for aggressive pituitary tumors and carcinomas. Presse Med 2016; 45:e211-6. [DOI: 10.1016/j.lpm.2016.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
27
|
Lin AL, Sum MW, DeAngelis LM. Is there a role for early chemotherapy in the management of pituitary adenomas? Neuro Oncol 2016; 18:1350-6. [PMID: 27106409 DOI: 10.1093/neuonc/now059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/16/2016] [Indexed: 12/25/2022] Open
Abstract
Pituitary adenomas are benign intracranial neoplasms that are frequently well-controlled with standard treatments that include surgical resection, radiotherapy, and agents that modulate hormonal excess. Unfortunately, a subset of patients remains uncontrolled or develops complications from these interventions. For these patients, chemotherapy is an additional treatment option that could improve outcomes. Temozolomide is an oral chemotherapy with a favorable side-effect profile that has shown activity against pituitary adenomas. Its non-overlapping toxicity and ability to induce rapid tumor regression renders it a potentially important adjunctive treatment. In patients with tumors that cannot be optimally addressed with standard treatments, there may be a role for early initiation of temozolomide.
Collapse
Affiliation(s)
- Andrew L Lin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.L.L., L.M.D.A.)
| | - Melissa W Sum
- Division of Endocrinology, Columbia University Medical Center, New York, New York (M.W.S.)
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.L.L., L.M.D.A.)
| |
Collapse
|
28
|
Auriemma RS, Grasso LFS, Pivonello R, Colao A. The safety of treatments for prolactinomas. Expert Opin Drug Saf 2016; 15:503-12. [DOI: 10.1517/14740338.2016.1151493] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Renata S. Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University ‘Federico II’, Naples, Italy
| | - Ludovica F. S. Grasso
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University ‘Federico II’, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University ‘Federico II’, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University ‘Federico II’, Naples, Italy
| |
Collapse
|
29
|
Kun Z, Yuling Y, Dongchun W, Bingbing X, Xiaoli L, Bin X. HIF-1α Inhibition Sensitized Pituitary Adenoma Cells to Temozolomide by Regulating Presenilin 1 Expression and Autophagy. Technol Cancer Res Treat 2015; 15:NP95-NP104. [PMID: 26647409 DOI: 10.1177/1533034615618834] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 10/06/2015] [Accepted: 10/29/2015] [Indexed: 11/17/2022] Open
Abstract
Pituitary adenomas usually develop temozolomide resistance, which could compromise the anticancer effects of temozolomide. Suppression of hypoxia-inducible factor 1α has been shown to sensitize glioblastoma cells to temozolomide treatment according to previous reports. However, whether and how the suppression of hypoxia-inducible factor 1α could sensitize pituitary adenomas to temozolomide treatment are still poorly understood. In the present study, using hypoxia-inducible factor 1α knockdown strategy, we demonstrated for the first time that hypoxia-inducible factor 1α knockdown could inhibit temozolomide-induced autophagy in rat pituitary adenoma GH3 cells and thus increase antitumor efficacy of temozolomide. Furthermore, we found hypoxia-inducible factor 1α knockdown could block autophagy process through neutralizing lysosomal pH value but not inhibiting autophagy induction. Finally, we found hypoxia-inducible factor 1α could regulate lysosomal pH value through regulating full length presenilin 1 expression, and exogenous reexpression of presenilin 1could restore lysosome acidic levels. Our data indicated hypoxia-inducible factor 1α knockdown could be a potential approach to improve the efficacy of temozolomide therapy for pituitary adenomas.
Collapse
Affiliation(s)
- Zhang Kun
- Department of Neurosurgery, Tangshan, China
| | | | | | | | - Li Xiaoli
- Department of Administration, Tangshan Worker Hospital, Tangshan, China
| | - Xu Bin
- Department of Neurosurgery, Hospital of Tangshan Iron and Steel Group Corporation, Tangshan, China
| |
Collapse
|
30
|
Ji Y, Vogel RI, Lou E. Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports. Neurooncol Pract 2015; 3:188-195. [PMID: 27551432 DOI: 10.1093/nop/npv059] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Pituitary carcinomas (PC) and atypical pituitary adenomas (APA) are rare variants of pituitary tumors for which no evidence-based treatment currently exists. We sought to determine whether temozolomide represents an effective chemotherapeutic option for patients with PC and APA. METHODS A systematic review was performed using all published cases of PC and APA treated with temozolomide, and for which information on treatment regimen, clinical response, and survival could be identified. The primary goal of this analysis was to describe overall survival and progression-free survival among PC and APA patients after temozolomide treatment. Secondary goals included assessment of response rate and biomarkers of response. RESULTS We identified 57 cases and obtained follow-up data on 54 patients (31 APA and 23 PC) for analysis. Estimates of 5-year progression-free survival and overall survival were 21.9% and 57.4% for patients with APA and 36.1% and 56.2% for patients with PC. Among those who responded to temozolomide, overall survival was marginally statistically significantly greater for patients on long-term temozolomide therapy compared with those who were not (5-year overall survival 91.7% vs 54.1%, P = .08); Progression-free survival results were similar but not statistically significant. The objective response rate was 48.4% for patients with APA and 65.2% for patients with PC. Stable disease occurred in 29% of APA and 17.4% of PC patients. Neither histology nor expression of Ki-67 correlated with response; however, negative O6-methylguanine-DNA methyltransferase staining was strongly related to response to temozolomide in patients with APA (P < .001). CONCLUSIONS Temozolomide is an effective treatment of both PC and APA, and long-term treatment can be considered for particularly aggressive cases.
Collapse
Affiliation(s)
- Yan Ji
- Division of Hematology, Oncology and Transplantation , University of Minnesota , Minneapolis, MN (Y.J., E.L.); Masonic Cancer Center, Biostatistics and Bioinformatics , University of Minnesota , Minneapolis, MN (R.I.V.)
| | - Rachel Isaksson Vogel
- Division of Hematology, Oncology and Transplantation , University of Minnesota , Minneapolis, MN (Y.J., E.L.); Masonic Cancer Center, Biostatistics and Bioinformatics , University of Minnesota , Minneapolis, MN (R.I.V.)
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation , University of Minnesota , Minneapolis, MN (Y.J., E.L.); Masonic Cancer Center, Biostatistics and Bioinformatics , University of Minnesota , Minneapolis, MN (R.I.V.)
| |
Collapse
|
31
|
Di Ieva A, Davidson JM, Syro LV, Rotondo F, Montoya JF, Horvath E, Cusimano MD, Kovacs K. Crooke's cell tumors of the pituitary. Neurosurgery 2015; 76:616-22. [PMID: 25635886 DOI: 10.1227/neu.0000000000000657] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Crooke's cell adenomas are a rare type of pituitary neoplasm. They produce adrenocorticotropic hormone causing Cushing's disease or may be endocrinologically silent. These tumors are usually invasive, may exhibit aggressive clinical behavior, and often recur with a low success of cure after reoperation and/or radiotherapy. Due to their rarity, they present great difficulties in assessing prognosis, treatment, and clinical management. Neurosurgeons and physicians dealing with pituitary adenomas diagnosed as Crooke's cell adenomas have to be aware of their potential clinical aggressiveness to plan strict follow-up of patients and eventual multimodality treatment. We review here the published cases of Crooke's cell tumors, as well as the clinical and histopathological characteristics of these unusual neoplasms.
Collapse
Affiliation(s)
- Antonio Di Ieva
- ‡Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; §Department of Neurosurgery, Hospital Pablo Tobon Uribe and Clinica Medellin, Medellin, Colombia; ¶Division of Pathology, Department of Laboratory Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; ‖Division of Endocrinology, Hospital Pablo Tobon Uribe and Universidad de Antioquia, Medellin, Colombia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Jiang XB, Hu B, He DS, Mao ZG, Wang X, Song BB, Zhu YH, Wang HJ. Expression profiling of O(6) methylguanine-DNA-methyl transferase in prolactinomas: a correlative study of promoter methylation and pathological features in 136 cases. BMC Cancer 2015; 15:644. [PMID: 26400193 PMCID: PMC4581044 DOI: 10.1186/s12885-015-1595-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 08/07/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Low-level expression of O(6) methylguanine-DNA-methyl transferase (MGMT) prolactinomas has been noted previously in case reports, although what modulates MGMT expression remains unclear. This study therefore aimed to delineate the factors regulating MGMT expression in prolactinomas. METHODS We retrospectively reviewed 136 prolactinoma patients who were treated in our center between January 2000 and September 2013. Expression of MGMT, Ki-67, and p53 protein were examined by immunohistochemical staining, and MGMT promoter methylation evaluated with methylation-specific PCR. RESULTS MGMT immunopositivity was <25 % in 106/136 tumor specimens (77.94 %). MGMT immunoexpression was positively correlated with age (r = 0.251, p = 0.003), but inversely correlated with p53 staining (r = -0.153, p = 0.021). Moreover, reduced MGMT expression was more frequent in atypical prolactinomas (p = 0.044). Methylated MGMT promoter was confirmed in 10/46 specimens (21.7 %), all of which had low level or absent MGMT staining. Both p53 protein (r = -0.33, p = 0.025) and promoter methylation (r = -0.331, p = 0.025) were negatively associated with MGMT expression. Multivariate logistic analysis indicated that age (odds ratio [OR] = 1.127. 95 % confidence interval [CI] 1.027-1.236, p = 0.012) and p53 (OR = 0.116. 95 % CI 0.018-0.761, p = 0.025) staining were independent determents of MGMT expression. CONCLUSIONS The majority of prolactinomas, especially atypical prolactinomas, showed low-level or no MGMT immunoexpression, providing a rationale for the utility of temozolomide as an alternative to managing prolactinomas. In summary, epigenetic and transcriptional regulation are involved in silencing MGMT expression.
Collapse
Affiliation(s)
- Xiao-Bing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-sen University, Haizhu, Yixian Rd, Guangdong, Guangzhou, China. .,Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China. .,Department of Neurosurgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| | - Bin Hu
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-sen University, Haizhu, Yixian Rd, Guangdong, Guangzhou, China. .,Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China.
| | - Dong-Sheng He
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-sen University, Haizhu, Yixian Rd, Guangdong, Guangzhou, China. .,Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China.
| | - Zhi-Gang Mao
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-sen University, Haizhu, Yixian Rd, Guangdong, Guangzhou, China. .,Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China.
| | - Xin Wang
- Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China. .,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Bing-Bing Song
- Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China. .,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yong-Hong Zhu
- Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China. .,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Hai-Jun Wang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-sen University, Haizhu, Yixian Rd, Guangdong, Guangzhou, China. .,Key Laboratory of Pituitary Adenoma in Guangdong Province, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Köchling M, Ewelt C, Fürtjes G, Peetz-Dienhart S, Koos B, Hasselblatt M, Paulus W, Stummer W, Brokinkel B. hTERT promoter methylation in pituitary adenomas. Brain Tumor Pathol 2015; 33:27-34. [PMID: 26390879 DOI: 10.1007/s10014-015-0230-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
Telomerase reverse transcriptase (TERT) expression is a hallmark in tumorigenesis and upregulated due to mutations and methylation of the human (h)TERT promoter. As mutations are rare but methylation is common in pituitary adenomas (PA), we determined promoter methylation and its clinical impact in 85 primary and 15 recurrent PA by methylation-specific PCR. 40 females (47%) and 45 males (53%) with a median age of 53 years harboring micro-, macro-, and giant adenomas in 12, 82, and 6% were included (prolactinomas, corticotroph, somatotroph, gonadotroph, thyreotroph, plurihormonal, and null cell adenomas in 11, 18, 10, 29, 1, 10, and 21%, respectively). In primary diagnosed tumors, methylation rate was 27% and higher in males than in females (40 vs. 13%, p = 0.001) after uni- and multivariate analyses. Methylation differed among PA subtypes (0-42%, p = n.s.) and was not significantly correlated with tumor size, cavernous sinus invasion, or serum hormone levels. Ki67 labeling index and recurrence (N = 16, 19%) were independent of methylation. In recurrent tumors, methylation was similar to primary PA (N = 5/15, 33%) and remained unchanged along follow-up. Thus, while being commonly observed in PA, hTERT promoter methylation is stable along follow-up and independent of most clinical variables, PA subtype, proliferation, and without prognostic value.
Collapse
Affiliation(s)
- Michaela Köchling
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, A1, 48149, Münster, Germany
| | - Christian Ewelt
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, A1, 48149, Münster, Germany
| | - Gina Fürtjes
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, A1, 48149, Münster, Germany
| | - Susanne Peetz-Dienhart
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Björn Koos
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, A1, 48149, Münster, Germany
| | - Benjamin Brokinkel
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, A1, 48149, Münster, Germany.
| |
Collapse
|
34
|
Zhao Y, Xiao Z, Chen W, Yang J, Li T, Fan B. Disulfiram sensitizes pituitary adenoma cells to temozolomide by regulating O6-methylguanine-DNA methyltransferase expression. Mol Med Rep 2015; 12:2313-22. [PMID: 25937029 DOI: 10.3892/mmr.2015.3664] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 03/24/2015] [Indexed: 11/06/2022] Open
Abstract
O6-methylguanine-DNA methyltransferase (MGMT) activity is responsible for temozolomide (TMZ) resistance in patients harboring aggressive pituitary adenomas. Recently, disulfiram (DSF) has been shown to induce the loss of MGMT protein and increase TMZ efficacy in glioblastoma cells, while CD133+ nestin+ cells isolated from the cell population have been implicated as pituitary adenoma stem-like cells. However, whether DSF is able to potentiate the cytotoxic effects of TMZ on human pituitary adenoma cells has not been investigated to date. In the present study, CD133+ nestin+ phenotype cells were isolated from primary cultured human pituitary adenoma cells using microbeads. It was found that DSF reduced MGMT protein expression and sensitized human pituitary adenoma cells and stem-like cells to TMZ in vitro, while the proteasome inhibitor PS-341 abrogated the inhibitory effect of DSF on MGMT in vitro. The sensitizing effect of DSF was also verified in primary cultured human pituitary adenoma cells in vivo. The results of the present study suggested that DSF can increase the efficacy of the anti-tumor effect of TMZ on human pituitary adenoma cells and CD133+ nestin+ stem like cells via the ubiquitin-proteasomal MGMT protein elimination route. DSF combined with TMZ may be an effective therapeutic strategy against aggressive pituitary adenomas.
Collapse
Affiliation(s)
- Yachao Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Zheng Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Wenna Chen
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Jinsheng Yang
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Tao Li
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Bo Fan
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
35
|
Liu JK, Patel J, Eloy JA. The role of temozolomide in the treatment of aggressive pituitary tumors. J Clin Neurosci 2015; 22:923-9. [PMID: 25772801 DOI: 10.1016/j.jocn.2014.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 01/09/2023]
Abstract
Pituitary tumors are amongst the most common intracranial neoplasms and are generally benign. However, some pituitary tumors exhibit clinically aggressive behavior that is characterized by tumor recurrence and continued progression despite repeated treatments with conventional surgical, radiation and medical therapies. More recently, temozolomide, a second generation oral alkylating agent, has shown therapeutic promise for aggressive pituitary adenomas and carcinomas with favorable clinical and radiographic responses. Temozolomide causes DNA damage by methylation of the O(6) position of guanine, which results in potent cytotoxic DNA adducts and consequently, tumor cell apoptosis. The degree of MGMT expression appears to be inversely related to therapeutic responsiveness to temozolomide with a significant number of temozolomide-sensitive pituitary tumors exhibiting low MGMT expression. The presence of high MGMT expression appears to mitigate the effectiveness of temozolomide and this has been used as a marker in several studies to predict the efficacy of temozolomide. Recent evidence also suggests that mutations in mismatch repair proteins such as MSH6 could render pituitary tumors resistant to temozolomide. In this article, the authors review the development of temozolomide, its biochemistry and interaction with O(6)-methylguanine-DNA methyltransferase (MGMT), its role in adjuvant treatment of aggressive pituitary neoplasms, and future works that could influence the efficacy of temozolomide therapy.
Collapse
Affiliation(s)
- James K Liu
- Center for Skull Base and Pituitary Surgery, Neurological Institute of New Jersey, Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Suite 8100, 90 Bergen Street, Newark, NJ 07103, USA; Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Newark, NJ, USA; Department of Otolaryngology-Head and Neck Surgery, Rutgers University, New Jersey Medical School, Newark, NJ, USA.
| | - Jimmy Patel
- Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Jean Anderson Eloy
- Center for Skull Base and Pituitary Surgery, Neurological Institute of New Jersey, Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Suite 8100, 90 Bergen Street, Newark, NJ 07103, USA; Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Newark, NJ, USA; Department of Otolaryngology-Head and Neck Surgery, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
36
|
Strowd RE, Salvatori R, Laterra JJ. Temozolomide retreatment in a recurrent prolactin-secreting pituitary adenoma: Hormonal and radiographic response. J Oncol Pharm Pract 2015; 22:517-22. [PMID: 25616657 DOI: 10.1177/1078155215569556] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Temozolomide is an oral alkylating agent with schedule-dependent antitumor activity against high-grade malignancies including high-grade glioma. Increasingly, reports have suggested that temozolomide may have activity as a salvage therapy for aggressive, recurrent pituitary adenomas or carcinomas that fail surgery, radiation and other pharmacotherapy. To our knowledge, temozolomide retreatment following initial responsiveness has not previously been demonstrated. CASE REPORT A woman was diagnosed with a prolactin-secreting pituitary adenoma in 1995 (age 44). Despite bromocriptine therapy, transphenoidal resection, radiotherapy, and cabergoline treatment she experienced continued clinico-radiographic progression, and temozolomide was initiated in 2011. She received three treatment cycles with rapid, dramatic clinico-radiographic response, and 99.3% reduction in serum prolactin. After three years of close observation, she developed recurrent radiographic progression and prolactin elevation. She was re-initiated on temozolomide, and after four cycles, clinical, radiographic and hormonal response was observed with a 92.2% reduction in serum prolactin. CONCLUSIONS/SUMMARY Temozolomide is an increasingly described treatment option for refractory pituitary adenomas and carcinomas. In the current report, we document rapid biochemical response following retreatment with temozolomide in aggressive pituitary adenoma. When "off label" salvage therapy with temozolomide is offered for patients with recurrent prolactinomas, retreatment at the time of recurrence can be considered.
Collapse
Affiliation(s)
- R E Strowd
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, USA
| | - R Salvatori
- Division of Endocrinology and Pituitary Center, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, USA
| | - J J Laterra
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, USA
| |
Collapse
|
37
|
Chatzellis E, Alexandraki KI, Androulakis II, Kaltsas G. Aggressive pituitary tumors. Neuroendocrinology 2015; 101:87-104. [PMID: 25571935 DOI: 10.1159/000371806] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/25/2014] [Indexed: 11/19/2022]
Abstract
Pituitary adenomas are common intracranial tumors that are mainly considered as benign. Rarely, these tumors can exhibit an aggressive behavior, characterized by gross invasion of the surrounding tissues, resistance to conventional treatment leading to early and frequent recurrences. Even more rarely, pituitary tumors can give rise to cerebrospinal or systemic metastases qualifying as pituitary carcinomas according to the latest WHO definition. In the same classification, a subset of tumors with relatively distinct histopathological features was identified and defined as atypical adenomas designated to follow a more aggressive clinical course. This classification, although clinically useful, does not provide an accurate correlation between histopathological findings and the clinical behavior of these tumors, neither is it adequate to convey the precise features of 'aggressive' tumors. Thus, 'aggressive' pituitary adenomas need to be properly defined with clinical, radiological, histological and molecular markers in order to identify patients at increased risk of early recurrence or subsequent tumor progression. At present, no single marker or classification system of pituitary tumor aggressiveness exists, and clinically useful information in the literature is insufficient to guide diagnostic and therapeutic decisions. Treatment of patients with aggressive pituitary tumors is challenging since conventional treatments often fail, necessitating multiple surgical procedures with additional radiotherapy. Although traditional chemotherapy applied in other neuroendocrine tumors has not been shown to be efficacious, newer agents, particularly temozolomide, have shown promising results and are currently used despite the lack of data from a randomized prospective trial. Molecular targeted therapies such as mTOR and epidermal growth factor inhibitors have also been applied and might prove to be useful in the management of these patients. In the present review, we provide information regarding the epidemiology and clinical, histopathological and molecular features of aggressive pituitary tumors using recent employed definitions. In addition, we review currently employed therapeutic means providing a therapeutic algorithm and highlight the need to identify more specific disease-related and prognostic markers and the necessity for central registration of these tumors.
Collapse
Affiliation(s)
- Eleftherios Chatzellis
- Endocrine Unit, Department of Pathophysiology, National University of Athens, Athens, Greece
| | | | | | | |
Collapse
|
38
|
Zacharia BE, Gulati AP, Bruce JN, Carminucci AS, Wardlaw SL, Siegelin M, Remotti H, Lignelli A, Fine RL. High response rates and prolonged survival in patients with corticotroph pituitary tumors and refractory Cushing disease from capecitabine and temozolomide (CAPTEM): a case series. Neurosurgery 2014; 74:E447-55; discussion E455. [PMID: 24226425 DOI: 10.1227/neu.0000000000000251] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND IMPORTANCE Rarely, corticotrophic pituitary tumors take on an aggressive form characterized by rapid growth, invasion into local structures, compression of cranial nerves, and possible spread to distant sites. When conventional surgery, radiation therapy, and hormones fail to control progression and symptoms, alternative therapies are needed. A novel chemotherapeutic regimen of capecitabine and temozolomide (CAPTEM), originally designed in our laboratory, demonstrated dramatic antineoplastic effects against corticotrophic pituitary tumors. CLINICAL PRESENTATION We present a case series of 4 patients with aggressive, adrenocorticotrophic hormone--producing pituitary tumors who had previously depleted all surgical, radiation, and hormonal therapies and were then treated with CAPTEM. Dramatic clinical improvements in neurological deficits and Cushing symptoms were evident in all patients after treatment was initiated. Confirmed by radiographic imaging, 2 of 4 patients demonstrated complete regression of disease, 1 patient had a 75% regression, and the fourth patient has ongoing stable disease for > 4.5 years at the time of this writing. Immunohistochemical analysis of patients' tumor samples showed low O-methyguanyl methyltransferase expression and adequate levels of mismatch repair enzymes (MLH-1, MSH-2, MSH-6, and PMS-2), which are important for the in vivo efficacy of CAPTEM. CONCLUSION This is the first report of prolonged antitumor response to and radiographic complete remissions as a result of CAPTEM in patients with aggressive pituitary tumors who had exhausted all other therapies.
Collapse
Affiliation(s)
- Brad E Zacharia
- *Department of Neurological Surgery, ‡Experimental Therapeutics Program, Department of Medicine, Division of Medical Oncology, Pancreas Center at Columbia, §Department of Medicine, Neuroendocrine Unit, and ¶Department of Pathology, Herbert Irving Comprehensive Cancer Center, New York Presbyterian Hospital-Columbia University Medical Center, New York, NY
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
An analysis of granulation patterns, MIB-1 proliferation indices and p53 expression in 101 patients with acromegaly. Acta Neurochir (Wien) 2014; 156:2221-30; discussion 2230. [PMID: 25238988 DOI: 10.1007/s00701-014-2230-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/03/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Despite the wide spectrum of adenoma behavior in patients with acromegaly, the ability of most pathological markers to predict clinical and radiological behavior remains controversial. The authors sought to comprehensively examine clinical and radiological correlates of growth hormone (GH)-secreting pituitary adenomas with regard to several commonly used immunocytochemical techniques in patients undergoing transsphenoidal surgery for acromegaly. METHOD We performed a retrospective review of histopathological findings in 101 surgically resected GH adenomas. Tumors were assessed radiologically for different patterns of extension. Each tumor specimen was subject to immunocytochemical analysis, including assessments of granulation patterns, MIB-1 labeling indices, prolactin cosecretion, p53 expression and mitotic activity. Endocrinological outcome was assessed in 93 patients, with remission defined by the 2010 consensus criteria. RESULTS Most tumors were macroadenomas and almost half were invasive. When compared to densely granulated tumors, sparsely granulated adenomas were associated with a younger age at presentation, higher preoperative IGF-1 levels, elevated MIB-1 index and pure GH immunostaining, but did not differ significantly in terms of extrasellar invasion or outcome. Increased mitotic activity and p53 expression were also associated with higher proliferation indices and a younger age at presentation. Mixed GH/prolactin tumors demonstrated significantly higher remission rates, independent of variations in extrasellar growth. MIB-1 indices did not correlate with the preoperative GH/IGF-1 levels, adenoma size or Knosp grade. CONCLUSION The pathobiology of acromegaly is complex, and the clinicoradiological significance of subtyping on the basis of the markers employed in this study is debatable. Further investigation of newer molecular markers is warranted.
Collapse
|
40
|
Hirohata T, Ishii Y, Matsuno A. Treatment of pituitary carcinomas and atypical pituitary adenomas: a review. Neurol Med Chir (Tokyo) 2014; 54:966-73. [PMID: 25446382 PMCID: PMC4533354 DOI: 10.2176/nmc.ra.2014-0178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atypical pituitary adenomas (APAs) are aggressive tumors, harboring a Ki-67 (MIB-1) staining index of 3% or more, and positive immunohistochemical staining for p53 protein, according to the World Health Organization (WHO) classification in 2004. Pituitary carcinomas (PC) usually develop from progressive APAs and predominantly consist of hormone-generating tumors, defined by the presence of disseminations in the cerebrospinal system or systemic metastases. Most of the cases with these malignant pituitary adenomas underwent surgeries, irradiations and adjuvant medical treatments, nevertheless, the therapies are mainly palliative. Recently, the efficacy of temozolomide (TMZ), an orally administered alkylating agent, has been reported as an alternative medical treatment. However, some recent studies have demonstrated a significant recurrence rate after effective response to TMZ. Further clinical and pathological researches of malignant pituitary adenomas will be required to improve the outcome of patients with these tumors.
Collapse
|
41
|
Abstract
The genetic mutations underlying familial syndromes that include pituitary tumors are rarely noted in the majority of sporadic adenohypophyseal adenomas. In contrast, epigenetic dysregulation is common, resulting in differential expression of cell cycle and apoptosis regulators, adhesion molecules, growth factors, and metabolic determinants of cell function. Here, we discuss the diagnostic and therapeutic implications of these findings as the landscape of pituitary tumor defects unfolds.
Collapse
Affiliation(s)
- Sylvia L Asa
- Department of Pathology, University Health Network, 200 Elizabeth Street, 11th floor, Toronto, Ontario, Canada, M5G 2C4,
| | | |
Collapse
|
42
|
Wang Y, Li J, Tohti M, Hu Y, Wang S, Li W, Lu Z, Ma C. The expression profile of Dopamine D2 receptor, MGMT and VEGF in different histological subtypes of pituitary adenomas: a study of 197 cases and indications for the medical therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:56. [PMID: 25027022 PMCID: PMC4223393 DOI: 10.1186/s13046-014-0056-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/27/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND To study the expression of D2R, MGMT and VEGF for clinical significance in pituitary adenomas, and to predict the potential curative medical therapy of dopamine agonists, temozolomide and bevacizumab on pituitary adenomas. METHODS Immunohistochemistry and western blot were performed to detect the expression of expression of D2R, MGMT and VEGF in pituitary adenoma tissue samples. The ratio of high expression of D2R, MGMT or VEGF in different subtypes of PA was compared by the use of chi-squared tests. The relationships between D2R, MGMT and VEGF expression were assessed by the Spearman rank correlation test. The association between their expression and clinical parameters was analyzed using a chi-squared test, or Fisher's exact probability test when appropriate. RESULTS The data showed that in 197 different histological subtypes of pituitary adenomas (PAs), 64.9% of them were D2R high expression, 86.3% were MGMT low expression and 58.9% were VEGF high expression. D2R high expression existed more frequently in PRL- and GH- secreting PAs. MGMT low expression existed in all PA subtypes. VEGF high expression existed more frequently in PRL, ACTH, FSH secreting and non-functioning PAs. The data of western blot also support the results. Spearman's rank correlation analysis showed that expression of MGMT was positively associated with D2R (r = 0.154, P = 0.031) and VEGF (r = 0.161, P = 0.024) in PAs, but no correlation was showed between D2R and VEGF expression (r = -0.025, P = 0.725 > 0.05). The association between their expression and clinical parameters was analyzed using a chi-squared test, or Fisher's exact probability test when appropriate, but the result showed no significant association. CONCLUSIONS PRL-and GH-secreting PAs exist high expression of D2R, responding to dopamine agonists; Most PAs exist low expression of MGMT and high expression of VEGF, TMZ or bevacizumab treatment could be applied under the premise of indications.
Collapse
|
43
|
Ferone D, Pivonello C, Vitale G, Zatelli MC, Colao A, Pivonello R. Molecular basis of pharmacological therapy in Cushing's disease. Endocrine 2014; 46:181-98. [PMID: 24272603 DOI: 10.1007/s12020-013-0098-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/19/2013] [Indexed: 01/16/2023]
Abstract
Cushing's disease (CD) is a severe endocrine condition caused by an adrenocorticotropin (ACTH)-producing pituitary adenoma that chronically stimulates adrenocortical cortisol production and with potentially serious complications if not or inadequately treated. Active CD may produce a fourfold increase in mortality and is associated with significant morbidities. Moreover, excess mortality risk may persist even after CD treatment. Although predictors of risk in treated CD are not fully understood, the importance of early recognition and adequate treatment is well established. Surgery with resection of a pituitary adenoma is still the first line therapy, being successful in about 60-70 % of patients; however, recurrence within 2-4 years may often occur. When surgery fails, medical treatment can reduce cortisol production and ameliorate clinical manifestations while more definitive therapy becomes effective. Compounds that target hypothalamic-pituitary axis, glucocorticoid synthesis or adrenocortical function are currently used to control the deleterious effects of chronic glucocorticoid excess. In this review we describe and analyze the molecular basis of the drugs targeting the disease at central level, suppressing ACTH secretion, as well as at peripheral level, acting as adrenal inhibitors, or glucocorticoid receptor antagonists. Understanding of the underlying molecular mechanisms in CD and of glucocorticoid biology should promote the development of new targeted and more successful therapies in the future. Indeed, most of the drugs discussed have been tested in limited clinical trials, but there is potential therapeutic benefit in compounds with better specificity for the class of receptors expressed by ACTH-secreting tumors. However, long-term follow-up with management of persistent comorbidities is needed even after successful treatment of CD.
Collapse
Affiliation(s)
- Diego Ferone
- Endocrinology, Department of Internal Medicine and Medical Specialties & Center of Excellence for Biomedical Research, IRCCS AOU San Martino-IST, University of Genova, Viale Benedetto XV, 6, 16132, Genoa, Italy,
| | | | | | | | | | | |
Collapse
|
44
|
Zhou Y, Zhang X, Klibanski A. Genetic and epigenetic mutations of tumor suppressive genes in sporadic pituitary adenoma. Mol Cell Endocrinol 2014; 386:16-33. [PMID: 24035864 PMCID: PMC3943596 DOI: 10.1016/j.mce.2013.09.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/03/2013] [Indexed: 12/28/2022]
Abstract
Human pituitary adenomas are the most common intracranial neoplasms. Approximately 5% of them are familial adenomas. Patients with familial tumors carry germline mutations in predisposition genes, including AIP, MEN1 and PRKAR1A. These mutations are extremely rare in sporadic pituitary adenomas, which therefore are caused by different mechanisms. Multiple tumor suppressive genes linked to sporadic tumors have been identified. Their inactivation is caused by epigenetic mechanisms, mainly promoter hypermethylation, and can be placed into two groups based on their functional interaction with tumor suppressors RB or p53. The RB group includes CDKN2A, CDKN2B, CDKN2C, RB1, BMP4, CDH1, CDH13, GADD45B and GADD45G; AIP and MEN1 genes also belong to this group. The p53 group includes MEG3, MGMT, PLAGL1, RASSF1, RASSF3 and SOCS1. We propose that the tumor suppression function of these genes is mainly mediated by the RB and p53 pathways. We also discuss possible tumor suppression mechanisms for individual genes.
Collapse
Affiliation(s)
- Yunli Zhou
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Xun Zhang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Anne Klibanski
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
45
|
Raverot G, Jouanneau E, Trouillas J. Management of endocrine disease: clinicopathological classification and molecular markers of pituitary tumours for personalized therapeutic strategies. Eur J Endocrinol 2014; 170:R121-32. [PMID: 24431196 DOI: 10.1530/eje-13-1031] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pituitary tumours, the most frequent intracranial tumour, are historically considered benign. However, various pieces of clinical evidence and recent advances in pathological and molecular analyses suggest the need to consider these tumours as more than an endocrinological disease, despite the low incidence of metastasis. Recently, we proposed a new prognostic clinicopathological classification of these pituitary tumours, according to the tumour size (micro, macro and giant), type (prolactin, GH, FSH/LH, ACTH and TSH) and grade (grade 1a, non-invasive; 1b, non-invasive and proliferative; 2a, invasive; 2b, invasive and proliferative and 3, metastatic). In addition to this classification, numerous molecular prognostic markers have been identified, allowing a better characterisation of tumour behaviour and prognosis. Moreover, clinical and preclinical studies have demonstrated that pituitary tumours could be treated by some chemotherapeutic drugs or new targeted therapies. Our improved classification of these tumours should now allow the identification of prognosis markers and help the clinician to propose personalised therapies to selected patients presenting tumours with a high risk of recurrence.
Collapse
Affiliation(s)
- Gerald Raverot
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team, Lyon F-69372, France
| | | | | |
Collapse
|
46
|
Arya S, Majaid MA, Shwetha SD, Sravani K, Arivazhagan A, Sampath S, Santosh V. Implications of MGMT methylation status in pituitary adenoma. Pathol Res Pract 2014; 210:407-11. [PMID: 24690322 DOI: 10.1016/j.prp.2014.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 09/04/2013] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Very little literature exists on frequency of MGMT methylation status in pituitary adenoma. We assessed the frequency of MGMT methylation and protein expression in pituitary adenoma and correlated with patients' age group and prognosis. METHODS Tumor tissues from 30 patients with pituitary adenoma were evaluated for MGMT promoter methylation status by methylation specific PCR method. All tumors were also immunostained with MIB-1, anti-p53 and anti-MGMT antibodies. RESULTS MGMT methylation status was noted in 40% of cases (7/20 non-functioning adenomas and 5/10 functioning adenomas). MGMT protein expression on IHC was noted in 72.2% of unmethylated tumors, while only 41.6% of methylated tumors demonstrated protein expression. The mean labeling index of MGMT protein was higher in unmethylated tumors as compared to the methylated group, though the difference was not statistically significant (18.6% vs 8.8%; p=0.131). Tumor regrowth occurred in four unmethylated tumors as compared to none in methylated group. Even though there was no correlation between patient age and MGMT methylation status (p=0.823), we noted that the frequency of methylation in middle age patients (between 30 and 59 yrs) was 64.7%, which was higher compared to other age groups. CONCLUSION This is the first study from India showing MGMT promoter methylation status with protein expression in pituitary adenoma. We noted that tumor regrowth was higher in unmethylated tumors.
Collapse
Affiliation(s)
- Sneha Arya
- Department of Neuropathology, National Institute of Mental Health and NeuroSciences, Bangalore, India
| | - M A Majaid
- Department of Neurosurgery, National Institute of Mental Health and NeuroSciences, Bangalore, India
| | - S D Shwetha
- Department of Neuropathology, National Institute of Mental Health and NeuroSciences, Bangalore, India
| | - K Sravani
- Department of Neuropathology, National Institute of Mental Health and NeuroSciences, Bangalore, India
| | - A Arivazhagan
- Department of Neurosurgery, National Institute of Mental Health and NeuroSciences, Bangalore, India.
| | - S Sampath
- Department of Neurosurgery, National Institute of Mental Health and NeuroSciences, Bangalore, India
| | - V Santosh
- Department of Neuropathology, National Institute of Mental Health and NeuroSciences, Bangalore, India
| |
Collapse
|
47
|
Abstract
The history of pituitary pathology is a long one that dates back to biblical times, but the last 25 years have represented an era of "coming of age." The role of the pituitary in health and disease was the subject of many studies over the last century. With the development of electron microscopy, immunoassays, and immunohistochemistry, the functional alterations associated with pituitary disease have been clarified. The additional information provided by molecular genetic studies has allowed progress in understanding the pathogenesis of pituitary disorders. Nevertheless, many questions remain to be answered. For example, pathologists cannot morphologically distinguish locally aggressive adenomas from carcinomas when tumor is confined to the sella. Sadly, basal cell carcinoma, the most common carcinoma of skin, usually causes less morbidity than pituitary adenomas, which occur in almost 20 % of the general population, can cause significant illness and even death, and yet are still classified as benign. The opportunity to increase awareness of the impact of these common lesions on quality of life is the current challenge for physicians and patients. We anticipate that ongoing multidisciplinary approaches to pituitary disease research will offer new insights into diseases arising from this fascinating organ.
Collapse
Affiliation(s)
- Sylvia L Asa
- Department of Pathology, Laboratory Medicine Program, University Health Network, 200 Elizabeth Street, 11th floor, Toronto, Ontario, Canada, M5G 2C4,
| | | |
Collapse
|
48
|
Kopczak A, Renner U, Karl Stalla G. Advances in understanding pituitary tumors. F1000PRIME REPORTS 2014; 6:5. [PMID: 24592317 PMCID: PMC3883424 DOI: 10.12703/p6-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Pituitary tumors are common in the general population. Since neuroimaging techniques have improved, pituitary tumors are more often diagnosed incidentally. About 16.7% of the general population show changes in the pituitary gland. Predominantly, pituitary tumors are benign pituitary adenomas. Pituitary carcinomas or aggressive pituitary tumors are extremely rare. They might develop from benign adenomas. New genetic and epigenetic abnormalities help us to understand pituitary tumorigenesis and might lead to therapeutical targeting drugs in the future. Macroadenomas (>1 cm) can lead to visual field disturbances, compression of cranial nerves, hypopituitarism, and infiltration of the cavernous sinuses. The functional status of the pituitary tumor is important. About half to one third of all pituitary tumors are non-functioning pituitary adenomas. The other pituitary tumors show a specific pattern of hormone secretion. About 25% to 41% of all pituitary tumors are prolactinomas, acromegaly with production of growth hormone represents 10% to 15% of adenomas, Cushing's disease with production of adrenocorticotropic hormone accounts for 10%, and other hormonal characteristics are less common. Transsphenoidal resection and total adenomectomy are desirable. Radiosurgery has enriched the surgical treatment options. Surgical treatment is the intervention of choice except for prolactinomas, where pharmaceutical treatment is recommended. Pharmaceutical treatment consists of dopamine agonists such as cabergoline and somatostatin analogues that include octreotide and pasireotide; retinoic acid is of theoretical interest while peroxisome proliferator-activated receptor-gamma-ligands are not clinically useful. In acromegaly, pegvisomant is a further treatment option. Temozolomide should be considered in aggressive pituitary tumors. In general, pharmaceutical options developed recently have extended the repertoire of treatment possibilities of pituitary tumors.
Collapse
Affiliation(s)
- Anna Kopczak
- Max Planck Institute of Psychiatry, Clinical Neurendocrinology GroupKraepelinstraße 2-10, 80804 MunichGermany
| | - Ulrich Renner
- Max Planck Institute of Psychiatry, Clinical Neurendocrinology GroupKraepelinstraße 2-10, 80804 MunichGermany
| | - Günter Karl Stalla
- Max Planck Institute of Psychiatry, Clinical Neurendocrinology GroupKraepelinstraße 2-10, 80804 MunichGermany
| |
Collapse
|
49
|
|
50
|
Foss CD, Dalton HJ, Monk BJ, Chase DM, Farley JH. Protein profiling of ovarian cancers by immunohistochemistry to identify potential target pathways. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2014; 1:4. [PMID: 27231557 PMCID: PMC4877732 DOI: 10.1186/2053-6844-1-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/15/2014] [Indexed: 11/21/2022]
Abstract
Background To determine the protein expression profile (PEP) of primary and recurrent ovarian cancer patients in order to predict therapeutic targets for chemotherapy. Methods Tissue samples were submitted for PEP in two formats, including formalin-fixed paraffin-embedded tissue for immunohistochemistry (IHC) and fresh frozen tissue for oligonucleotide microarray (MA) gene expression assays. Specimens were analyzed for 18 protein markers and 88 MA genes. A series of Generalized Linear Models (GLM) was used to predict the proportion of positive results by histology for each biomarker. Results Four hundred and twenty-eight specimens were analyzed for IHC and 67 specimens for MA analysis. The majority of specimens, 82%, were serous histology and 35.3% of specimens were poorly differentiated. Sixty percent of specimens were advanced stage, 62% were from a primary diagnosis, and 53% were obtained from a metastatic site. BCRP, ER, MGMT, and RRM1 proteins were overexpressed in 85%, 47%, 93%, and 47% of serous carcinomas, respectively. The MGMT and RRM1 biomarkers were significantly overexpressed in serous (p < .001) and endometrioid (p = .01) histologies when compared to clear cell histology. MGMT was significantly elevated in 93% of serous and endometrioid samples, compared to 62% of samples with clear cell histology. Those proteins most often underexpressed included Her2/neu, SPARC, and c-kit, seen in less than 1%, 4%, and 5% of specimens, respectively. Conclusions PEP is a reliable and effective way of analyzing ovarian cancer specimens. PEP target identification does not appear to vary significantly with site evaluated, ovarian or other abdominal pelvic tissue, or primary versus recurrent disease. Variability in the expression of drug targets, including BCRP, ER, MGMT, and RRM1 could impact decision making pertaining to which therapeutic strategies carry the best chances for controlling disease. Electronic supplementary material The online version of this article (doi:10.1186/2053-6844-1-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cassandra D Foss
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arizona Cancer Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA ; Creighton University School of Medicine at Dignity Health St. Joseph's Hospital and Medical Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA
| | - Heather J Dalton
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Bradley J Monk
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arizona Cancer Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA ; Creighton University School of Medicine at Dignity Health St. Joseph's Hospital and Medical Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA
| | - Dana M Chase
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arizona Cancer Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA ; Creighton University School of Medicine at Dignity Health St. Joseph's Hospital and Medical Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA
| | - John H Farley
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arizona Cancer Center, 500 W. Thomas Road, Suite 600, Phoenix, AZ 85013 USA
| |
Collapse
|