1
|
Terry M, Nguyen MP, Tang V, Guney E, Bharani KL, Dahiya S, Choutka O, Borys E, Reis G, Blevins L, Aghi MK, Kunwar S, DeGroot J, Raleigh DR, Pekmezci M, Bollen AW, Cha S, Joseph NM, Perry A. High-Grade Progression, Sarcomatous Transformation, and/or Metastasis of Pituitary Neuroendocrine Neoplasms (PitNENs): The UCSF Experience. Endocr Pathol 2024:10.1007/s12022-024-09829-w. [PMID: 39388031 DOI: 10.1007/s12022-024-09829-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Pituitary neuroendocrine tumors (PitNET) that metastasize comprise ~ 0.2% of adenohypophyseal tumors are aggressive and are challenging to treat. However, many non-metastatic tumors are also aggressive. Herein, we review 21 specimens from 13 patients at UCSF with metastatic PitNETs (CSF or systemic, N = 7 patients), high-grade pituitary neuroendocrine neoplasms (HG-PitNEN, N = 4 patients), and/or PitNETs with sarcomatous transformation (PitNET-ST, N = 5 patients). We subtyped cases using the World Health Organization (WHO) and International Agency for Research on Cancer (IARC) criteria for neuroendocrine neoplasms (NENs). Lineage subtypes included acidophil stem cell, null cell, thyrotroph, corticotroph, lactotroph, and gonadotroph tumors. The median Ki-67 labeling index was 25% (range 5-70%). Lack of p16 was seen in 3 cases, with overexpression in 2. Strong diffuse p53 immunopositivity was present in 3 specimens from 2 patients. Loss of Rb expression was seen in 2 cases, with ATRX loss in one. Molecular analysis in 4 tumors variably revealed TERT alterations, homozygous CDKN2A deletion, aneuploidy, and mutations in PTEN, TP53, PDGFRB, and/or PIK3CA. Eight patients (62%) died of disease, 4 were alive at the last follow-up, and 1 was lost to the follow-up. All primary tumors had worrisome features, including aggressive lineage subtype, high mitotic count, and/or high Ki-67 indices. Additional evidence of high-grade progression included immunohistochemical loss of neuroendocrine, transcription factor, and/or hormone markers. We conclude that metastatic PitNET is not the only high-grade form of pituitary NEN. If further confirmed, these histopathologic and/or molecular features could provide advanced warning of biological aggressiveness and be applied towards a future grading scheme.
Collapse
Affiliation(s)
- Merryl Terry
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Minh P Nguyen
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Vivian Tang
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Ekin Guney
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | | | - Sonika Dahiya
- Department of Pathology, Washington University, St. Louis, MO, USA
| | - Ondrej Choutka
- Department of Neurosurgery, Saint Alphonsus Neuroscience Institute, Boise, ID, USA
| | - Ewa Borys
- Department of Pathology, Loyola University, Chicago, IL, USA
| | - Gerald Reis
- Department of Pathology, Memorial Healthcare System, Hollywood, FL, USA
| | - Lewis Blevins
- Department of Endocrinology, University of California San Francisco, San Francisco, CA, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sandeep Kunwar
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - John DeGroot
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - David R Raleigh
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Melike Pekmezci
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Andrew W Bollen
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Nancy M Joseph
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Arie Perry
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Kim YS, Ahn S, Lee YS, Jeun SS, Park JS. Clinicopathological analysis of non-functioning pituitary adenomas (PAs) according to the 2022 WHO classification. Pituitary 2024; 27:665-672. [PMID: 38896347 DOI: 10.1007/s11102-024-01414-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE For asymptomatic non-functioning pituitary adenomas (NFPAs), conservative approaches such as observation are preferred. However, some NFPAs exhibit poor prognoses. Thus, the purpose of this study was to investigate clinicopathological characteristics of tumors for identifying those with unfavorable prognoses. METHODS A total of 125 patients with NFPAs who underwent surgery between November 2017 and December 2022 at our institution were retrospectively analyzed. Clinical, radiological, and pathological data, including hormone profiles, tumor size, presence of cavernous sinus invasion, and Ki-67 index levels, were reviewed. High-risk PAs were identified according to 2022 WHO criteria. Statistical analyses including Kaplan-Meier survival analysis and Cox regression were performed to evaluate factors associated with tumor progression or recurrence. RESULTS A high-risk group demonstrated a significantly higher rate of tumor progression/recurrence than a low-risk group (p-value = 0.004). In multivariate analysis, the high-risk group at the time of diagnosis remained as an independent prognostic factor for NFPAs (p-value = 0.0148). The high-risk group also had a higher percentage of younger patients (80.0% in the high-risk group vs. 62.2% in the low-risk group, p-value = 0.016) and female patients (91.4% vs. 34.4%, p< 0.001). The presence of cavernous sinus invasion and higher Ki-67 index levels were more commonly observed in the high-risk group, although these factors did not significantly impact the overall prognosis. CONCLUSION Our findings indicate that patients with high-risk NFPAs have a more aggressive disease course and a higher rate of progression or recurrence. This high-risk group has higher prevalence of younger and female patients. They may benefit from closer monitoring and possibly more aggressive treatment approaches.
Collapse
Affiliation(s)
- Yeo Song Kim
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn-Soo Lee
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae-Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Zhang X, Chen Y, Yu Y, Li J. Diagnosis and Management of Aggressive/Refractory Growth Hormone-Secreting Pituitary Neuroendocrine Tumors. Int J Endocrinol 2024; 2024:5085905. [PMID: 39224564 PMCID: PMC11368557 DOI: 10.1155/2024/5085905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The majority of acromegaly and gigantism are caused by growth hormone-secreting pituitary neuroendocrine tumors (PitNETs). Most cases can be cured or controlled by surgery, medical therapy, and/or radiotherapy. However, a few of these tumors are resistant to traditional therapy and always have a poor prognosis. The title aggressive/refractory is used to differentiate them from pituitary carcinomas. To date, there is no definitive conclusion on how to diagnose aggressive/refractory growth hormone-secreting PitNETs, which may have slowed the process of exploring new therapeutical strategies. We summarized the literature described diagnosis and treatment of the disease. Potential disease markers and prospective therapies were also included.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Yerong Yu
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Jianwei Li
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Namiot ED, Zembatov GM, Tregub PP. Insights into brain tumor diagnosis: exploring in situ hybridization techniques. Front Neurol 2024; 15:1393572. [PMID: 39022728 PMCID: PMC11252041 DOI: 10.3389/fneur.2024.1393572] [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: 02/29/2024] [Accepted: 05/31/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives Diagnosing brain tumors is critical due to their complex nature. This review explores the potential of in situ hybridization for diagnosing brain neoplasms, examining their attributes and applications in neurology and oncology. Methods The review surveys literature and cross-references findings with the OMIM database, examining 513 records. It pinpoints mutations suitable for in situ hybridization and identifies common chromosomal and gene anomalies in brain tumors. Emphasis is placed on mutations' clinical implications, including prognosis and drug sensitivity. Results Amplifications in EGFR, MDM2, and MDM4, along with Y chromosome loss, chromosome 7 polysomy, and deletions of PTEN, CDKN2/p16, TP53, and DMBT1, correlate with poor prognosis in glioma patients. Protective genetic changes in glioma include increased expression of ADGRB3/1, IL12B, DYRKA1, VEGFC, LRRC4, and BMP4. Elevated MMP24 expression worsens prognosis in glioma, oligodendroglioma, and meningioma patients. Meningioma exhibits common chromosomal anomalies like loss of chromosomes 1, 9, 17, and 22, with specific genes implicated in their development. Main occurrences in medulloblastoma include the formation of isochromosome 17q and SHH signaling pathway disruption. Increased expression of BARHL1 is associated with prolonged survival. Adenomas mutations were reviewed with a focus on adenoma-carcinoma transition and different subtypes, with MMP9 identified as the main metalloprotease implicated in tumor progression. Discussion Molecular-genetic diagnostics for common brain tumors involve diverse genetic anomalies. In situ hybridization shows promise for diagnosing and prognosticating tumors. Detecting tumor-specific alterations is vital for prognosis and treatment. However, many mutations require other methods, hindering in situ hybridization from becoming the primary diagnostic method.
Collapse
Affiliation(s)
- E. D. Namiot
- Department of Pathophysiology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - G. M. Zembatov
- Department of Pathophysiology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - P. P. Tregub
- Department of Pathophysiology, First Moscow State Medical University (Sechenov University), Moscow, Russia
- Brain Research Department, Federal State Scientific Center of Neurology, Moscow, Russia
- Scientific and Educational Resource Center, Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
5
|
Пронин ВС, Анциферов МБ, Алексеева ТМ, Пронин ЕВ, Лапшина АМ, Урусова ЛС. [Unification of pathomorphological examination of patients with neuroendocrine tumors of the pituitary gland. Controversial issues of the new classification]. PROBLEMY ENDOKRINOLOGII 2023; 70:31-45. [PMID: 39069771 PMCID: PMC11334236 DOI: 10.14341/probl13376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 07/30/2024]
Abstract
The progressive improvement of the classification using modern analytical methods is an essential tool for the development of precise and personalized approaches to the treatment of pituitary adenomas. In recent years, endocrinologists have witnessed evolutionary changes that have occurred in the histopathological identification of pituitary neoplasms, revealing new possibilities for studying tumorigenesis and predicting biological behavior.The paper considers the historical aspects of the gradual improvement of the classification of pituitary adenomas, as well as the new international 2022 WHO classification, according to which pituitary adenomas are included in the list of neuroendocrine tumors (PitNETs) to reflect the biological aggressiveness of some non-metastatic pituitary adenomas. The characteristics of pituitary adenoma are presented, as well as a list of histological subtypes of aggressive neuroendocrine tumors of the pituitary gland, marked by the main potentials for invasive growth, an increased risk of recurrence and a negative clinical prognosis.The expediency of changing the definition of «pituitary adenoma» to «neuroendocrine tumor» is discussed. It is emphasized that the introduction of a unified clinical, laboratory and morphological protocol into national clinical practice will help provide comparable comparative studies on the prognosis of the disease and the effectiveness of secondary therapy and also contribute to adequate management of potentially aggressive PitNETs.
Collapse
Affiliation(s)
- В. С. Пронин
- Российская медицинская академия непрерывного профессионального образования;
Эндокринологический диспансер Департамента здравоохранения города Москвы
| | - М. Б. Анциферов
- Российская медицинская академия непрерывного профессионального образования;
Эндокринологический диспансер Департамента здравоохранения города Москвы
| | - Т. М. Алексеева
- Эндокринологический диспансер Департамента здравоохранения города Москвы
| | - Е. В. Пронин
- Эндокринологический диспансер Департамента здравоохранения города Москвы
| | - А. М. Лапшина
- Национальный медицинский исследовательский центр эндокринологии
| | - Л. С. Урусова
- Национальный медицинский исследовательский центр эндокринологии
| |
Collapse
|
6
|
Zhao J, Ji C, Cheng H, Ye Z, Yao B, Shen M, Shou X, Zhou X, Ye H, Zhang Z, Chen H, Wang Y, He F, Zhao Y, Gong W, Zhang Q, Qiao N. Digital image analysis allows objective stratification of patients with silent PIT1-lineage pituitary neuroendocrine tumors. J Pathol Clin Res 2023; 9:488-497. [PMID: 37661840 PMCID: PMC10556262 DOI: 10.1002/cjp2.340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/01/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023]
Abstract
Studies describing the clinical presentation and prognosis of patients with silent PIT1 (pituitary specific transcription factor)-lineage pituitary neuroendocrine tumors (PitNETs) are rare. We identified patients with positive PIT1 tumor staining but without evidence of hormone hypersecretion at a tertiary center. Clusters were obtained according to cell morphology and immunostaining from each patient's digitally segmented whole slide image. We compared the clinical presentations, radiological features, and prognoses of the different clusters. We identified 146 patients (68 male, 42.9 ± 14.1 years old) with silent PIT1-lineage PitNETs. Morphology clustering suggested that tumors with large nuclei and apparent eccentricity were associated with a higher proportion of aggressiveness and a higher hazard of recurrence [hazard ratio (HR): 2.64, (95% CI, 1.06-6.55), p = 0.037]. Immunohistochemical clustering suggested that tumors with thyroid stimulating hormone (TSH) staining or all negative PIT1-lineage hormones were associated with a higher proportion of aggressiveness and a higher risk of recurrence [HR: 12.4, (95% CI, 1.60-93.5), p = 0.015]. We obtained three-tier risk profiles by combining morphological and immunohistochemical clustering. Patients with the high-risk profile presented the highest recurrence rate compared with those in the medium-risk and low-risk profiles [HR: 3.54, (95% CI, 1.40-8.93), p = 0.002]. In conclusion, digital image analysis based on cell morphology and immunohistochemical staining allows objective stratification of patients with silent PIT1-lineage tumors. Typical morphological characteristics of high-risk tumors are large tumor nuclei and high eccentricity, and typical immunostaining characteristics are TSH staining or negative staining for all PIT1-lineage hormones.
Collapse
Affiliation(s)
- Jiangyan Zhao
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
| | - Chenxing Ji
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Haixia Cheng
- Department of PathologyHuashan HospitalShanghaiPR China
| | - Zhen Ye
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Boyuan Yao
- Fudan University Graduate SchoolShanghaiPR China
| | - Ming Shen
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Xuefei Shou
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Xiang Zhou
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Hongying Ye
- Department of EndocrinologyHuashan HospitalShanghaiPR China
| | - Zhaoyun Zhang
- Department of EndocrinologyHuashan HospitalShanghaiPR China
| | - Hong Chen
- Department of PathologyHuashan HospitalShanghaiPR China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Fuchu He
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- State Key Laboratory of Proteomics, Beijing Proteome Research CenterNational Center for Protein SciencesBeijingPR China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Wei Gong
- Department of EndocrinologyHuashan HospitalShanghaiPR China
| | - Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| | - Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical SciencesFudan UniversityShanghaiPR China
- National Center for Neurological DisordersShanghaiPR China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiPR China
- Neurosurgical Institute of Fudan UniversityShanghaiPR China
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiPR China
| |
Collapse
|
7
|
Shi M, Song Y, Zhang Y, Li L, Yu J, Hou A, Han S. PD-L1 and tumor-infiltrating CD8 + lymphocytes are correlated with clinical characteristics in pediatric and adolescent pituitary adenomas. Front Endocrinol (Lausanne) 2023; 14:1151714. [PMID: 37424874 PMCID: PMC10323746 DOI: 10.3389/fendo.2023.1151714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
Objective To investigate the levels of tumor-infiltrating CD8+ lymphocytes (CD8+ TILs) and the expression of programmed cell death receptor ligand 1 (PD-L1) in the tumor microenvironment (TME) of pediatric and adolescent pituitary adenomas (PAPAs) and analyze the correlation between their levels and the clinical characteristics. Methods A series of 43 PAPAs cases were enrolled over a period of 5 years. To compare the TME of PAPAs and adult PAs, 43 PAPAs cases were matched with 60 adult PAs cases (30 cases were between 20 and 40 years old, and 30 cases were older than 40 years) for main clinical characteristics. The expression of immune markers in PAPAs was detected by immunohistochemistry, and their correlation with the clinical outcomes was analyzed using statistical methods. Results In the PAPAs group, CD8+ TILs level was significantly lower (3.4 (5.7) vs. 6.1 (8.5), p = 0.001), and PD-L1 expression (0.040 (0.022) vs. 0.024 (0.024), p < 0.0001) was significantly higher as compared with the older group. The level of CD8+ TILs was negatively correlated with the expression of PD-L1 (r = -0.312, p = 0.042). Moreover, CD8+ TILs and PD-L1 levels were associated with Hardy (CD8, p = 0.014; PD-L1, p = 0.018) and Knosp (CD8, p = 0.02; PD-L1, p = 0.017) classification. CD8+ TILs level was associated with high-risk adenomas (p = 0.015), and it was associated with the recurrence of PAPAs (HR = 0.047, 95% CI 0.003-0.632, p = 0.021). Conclusion Compared with the TME in adult PAs, the TME in PAPAs was found to express a significantly altered level of CD8+ TILs and PD-L1. In PAPAs, CD8+ TILs and PD-L1 levels were associated with clinical characteristics.
Collapse
Affiliation(s)
- Mengwu Shi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Longjie Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Juanhan Yu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Kober P, Rusetska N, Mossakowska BJ, Maksymowicz M, Pękul M, Zieliński G, Styk A, Kunicki J, Działach Ł, Witek P, Bujko M. The expression of glucocorticoid and mineralocorticoid receptors in pituitary tumors causing Cushing's disease and silent corticotroph tumors. Front Endocrinol (Lausanne) 2023; 14:1124646. [PMID: 37065760 PMCID: PMC10090509 DOI: 10.3389/fendo.2023.1124646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
OBJECTIVE Pituitary neuroendocrine corticotroph tumors commonly cause Cushing's disease (CD) that results from increased adrenocorticotropic hormone (ACTH) secretion by the pituitary tumor and consequent increase of cortisol levels in blood. However, in some patients, corticotroph tumors remain clinically non-functioning. Cortisol secretion is regulated by the hypothalamic-pituitary-adrenal axis and includes a negative feedback between cortisol and ACTH secretion. Glucocorticoids reduce ACTH level both by hypothalamic regulation and acting on corticotrophs via glucocorticoid (GR) and mineralocorticoid (MR) receptors. The aim of the study was to determine the role of GR and MR expression at mRNA and protein levels in both functioning and silent corticotroph tumors. METHODS Ninety-five patients were enrolled, including 70 with CD and 25 with silent corticotroph tumors. Gene expression levels of NR3C1 and NR3C2 coding for GR and MR, respectively, were determined with qRT-PCR in the two tumor types. GR and MR protein abundance was assessed with immunohistochemistry. RESULTS Both GR and MR were expressed in corticotroph tumors. Correlation between NR3C1 and NR3C2 expression levels was observed. NR3C1 expression was higher in silent than in functioning tumors. In CD patients NR3C1 and NR3C2 levels were negatively correlated with morning plasma ACTH levels and tumor size. Higher NR3C2 was confirmed in patients with remission after surgery and in densely granulated tumors. Expression of both genes and GR protein was higher in USP8-mutated tumors. Similar relationship between USP8 mutations and expression levels were observed in analysis of silent tumors that also revealed a negative correlation between GR and tumor size and higher NR3C1 expression in densely granulated tumors. CONCLUSIONS Although the associations between gene/protein expression and patients clinical features are not strong, they consistently show an evident trend in which higher receptor expression corresponds to more favorable clinical characteristics.
Collapse
Affiliation(s)
- Paulina Kober
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Natalia Rusetska
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata J. Mossakowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Maksymowicz
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Monika Pękul
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Grzegorz Zieliński
- Department of Neurosurgery, Military Institute of Medicine, Warsaw, Poland
| | - Andrzej Styk
- Department of Neurosurgery, Military Institute of Medicine, Warsaw, Poland
| | - Jacek Kunicki
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Łukasz Działach
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Witek
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- *Correspondence: Mateusz Bujko,
| |
Collapse
|
9
|
Transcriptomic Classification of Pituitary Neuroendocrine Tumors Causing Acromegaly. Cells 2022; 11:cells11233846. [PMID: 36497102 PMCID: PMC9738119 DOI: 10.3390/cells11233846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Acromegaly results from growth hormone hypersecretion, predominantly caused by a somatotroph pituitary neuroendocrine tumor (PitNET). Acromegaly-causing tumors are histologically diverse. Our aim was to determine transcriptomic profiles of various somatotroph PitNETs and to evaluate clinical implication of differential gene expression. A total of 48 tumors were subjected to RNA sequencing, while expression of selected genes was assessed in 134 tumors with qRT-PCR. Whole-transcriptome analysis revealed three transcriptomic groups of somatotroph PitNETs. They differ in expression of numerous genes including those involved in growth hormone secretion and known prognostic genes. Transcriptomic subgroups can be distinguished by determining the expression of marker genes. Analysis of the entire cohort of patients confirmed differences between molecular subtypes of tumors. Transcriptomic group 1 includes ~20% of acromegaly patients with GNAS mutations-negative, mainly densely granulated tumors that co-express GIPR and NR5A1 (SF-1). SF-1 expression was verified with immunohistochemistry. Transcriptomic group 2 tumors are the most common (46%) and include mainly GNAS-mutated, densely granulated somatotroph and mixed PitNETs. They have a smaller size and express favorable prognosis-related genes. Transcriptomic group 3 includes predominantly sparsely granulated somatotroph PitNETs with low GNAS mutations frequency causing ~35% of acromegaly. Ghrelin signaling is implicated in their pathogenesis. They have an unfavorable gene expression profile and higher invasive growth rate.
Collapse
|
10
|
Li Z, Chen Y, Yao X, Liu Q, Zhu H, Zhang Y, Feng J, Gao H. The Integrated Stress Response Is Tumorigenic and Constitutes a Therapeutic Liability in Somatotroph Adenomas. Int J Mol Sci 2022; 23:ijms232113067. [PMID: 36361871 PMCID: PMC9653568 DOI: 10.3390/ijms232113067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Somatotroph adenomas are the leading cause of acromegaly, with the nearly sparsely granulated somatotroph subtype belonging to high-risk adenomas, and they are less responsive to medical treatment. The integrated stress response (ISR) is an essential stress-support pathway increasingly recognized as a determinant of tumorigenesis. In this study, we identified the characteristic profiling of the integrated stress response in translocation and translation initiation factor activity in somatotroph adenomas, normal pituitary, or other adenoma subtypes through proteomics. Immunohistochemistry exhibited the differential significance and the priority of eukaryotic translation initiation factor 2β (EIF2β) in somatotroph adenomas compared with gonadotroph and corticotroph adenomas. Differentially expressed genes based on the level of EIF2β in somatotroph adenomas were revealed. MetaSape pathways showed that EIF2β was involved in regulating growth and cell activation, immune system, and extracellular matrix organization processes. The correlation analysis showed Spearman correlation coefficients of r = 0.611 (p < 0.001) for EIF2β and eukaryotic translation initiation factor 2 alpha kinase 1 (HRI), r = 0.765 (p < 0.001) for eukaryotic translation initiation factor 2 alpha kinase 2 (PKR), r = 0.813 (p < 0.001) for eukaryotic translation initiation factor 2 alpha kinase 3 (PERK), r = 0.728 (p < 0.001) for GCN2, and r = 0.732 (p < 0.001) for signal transducer and activator of transcription 3 (STAT3). Furthermore, the invasive potential in patients with a high EIF2β was greater than that in patients with a low EIF2β (7/10 vs. 4/18, p = 0.038), with a lower immune-cell infiltration probability (p < 0.05). The ESTIMATE algorithm showed that the levels of activation of the EIF2 pathway were negatively correlated with the immune score in somatotroph adenomas (p < 0.001). In in vitro experiments, the knockdown of EIF2β changed the phenotype of somatotroph adenomas, including cell proliferation, migration, and the secretion ability of growth hormone/insulin-like growth factor-1. In this study, we demonstrate that the ISR is pivotal in somatotroph adenomas and provide a rationale for implementing ISR-based regimens in future treatment strategies.
Collapse
Affiliation(s)
- Zhenye Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yiyuan Chen
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Xiaohui Yao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Shanxi Provincial People’s Hospital, Taiyuan 030000, China
| | - Qian Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Haibo Zhu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yazhuo Zhang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Jie Feng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Correspondence: (J.F.); (H.G.)
| | - Hua Gao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Key Laboratory of Central Nervous System Injury Research, Beijing 100070, China
- Correspondence: (J.F.); (H.G.)
| |
Collapse
|
11
|
Honegger J, Nasi-Kordhishti I. Surgery and perioperative management of patients with Cushing's disease. J Neuroendocrinol 2022; 34:e13177. [PMID: 35980172 DOI: 10.1111/jne.13177] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/09/2022] [Accepted: 05/31/2022] [Indexed: 11/29/2022]
Abstract
Transsphenoidal surgery (TSS) is the initial treatment modality of first choice in Cushing's disease (CD). With microscopic TSS and endoscopic TSS, two operative techniques with equally favourable remission rates and operative morbidity are available. On average, remission is achieved with primary TSS in 80% of patients with microadenomas and 60% of patients with macroadenomas. The current literature indicates that remission rates in repeat TSS for microadenomas can also exceed 70%. Experience with TSS in CD plays an important role in the success rate and centralization in Pituitary Centres of Excellence has been proposed. Microadenoma stage, imaging-visible adenoma, confirmation of ACTH-positive adenoma on histopathology and noninvasiveness are positive predictors for postoperative remission. In postoperative management, a steroid-sparing protocol with early postoperative assessment of remission status is recommended. Convincing evidence suggests that prolonged postoperative prophylactic antithrombotic measures can significantly reduce the risk of postoperative thromboembolic events in CD. Prevention or successful treatment of cortisol withdrawal syndrome remains an unsolved issue that should be a focus of future research. The further development and broad availability of functional imaging hold promise for improved preoperative detection of microadenomas. Intraoperative identification of microadenomas by specific fluorescent targeting could be a promising future avenue for the treatment of patients with negative imaging.
Collapse
Affiliation(s)
- Juergen Honegger
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
12
|
Du P, Wu X, Lv K, Xiong J, Geng D. A Pituitary Carcinoma Patient With Cerebrospinal Fluid Dissemination Showing a Good Response to Temozolomide Combined With Whole-Brain and Spinal Cord Radiotherapy: A Case Report and Literature Review. Front Oncol 2022; 12:890458. [PMID: 35903687 PMCID: PMC9321396 DOI: 10.3389/fonc.2022.890458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/20/2022] [Indexed: 12/03/2022] Open
Abstract
Pituitary carcinoma (PC) is extremely rare, with its incidence only accounting for 0.1%-0.2% of pituitary tumor (PT). Existing histological features, including invasiveness, cellular pleomorphism, nuclear atypia, mitosis, necrosis, etc., can be observed in pituitary adenoma (PA), invasive PA (IPA) and PC. Invasion is not the basis for the diagnosis of PC. The diagnosis of PC is often determined after the metastases are found, hence early diagnosis is extraordinarily difficult. Owing to the conventional treatment for PC may not be effective, a large portion of patients survived less than one year after diagnosis. Therefore, it is of great significance to find an efficacious treatment for PC. We report a rare case of sparsely granulated somatotroph carcinoma with cerebrospinal fluid dissemination showing a favorable treatment response to temozolomide (TMZ) combined with whole-brain and spinal cord radiotherapy.
Collapse
Affiliation(s)
- Peng Du
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Center for Shanghai Intelligent Imaging for Critical Brain Diseases Engineering and Technology Research, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuefan Wu
- Department of Radiology, Shanghai Gamma Hospital, Huashan Hospital, Fudan University, Shanghai, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Center for Shanghai Intelligent Imaging for Critical Brain Diseases Engineering and Technology Research, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Daoying Geng,
| |
Collapse
|
13
|
Difference in miRNA Expression in Functioning and Silent Corticotroph Pituitary Adenomas Indicates the Role of miRNA in the Regulation of Corticosteroid Receptors. Int J Mol Sci 2022; 23:ijms23052867. [PMID: 35270010 PMCID: PMC8911444 DOI: 10.3390/ijms23052867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Corticotroph pituitary adenomas commonly cause Cushing’s disease (CD), but some of them are clinically silent. The reason why they do not cause endocrinological symptoms remains unclear. We used data from small RNA sequencing in adenomas causing CD (n = 28) and silent ones (n = 20) to explore the role of miRNA in hormone secretion and clinical status of the tumors. By comparing miRNA profiles, we identified 19 miRNAs differentially expressed in clinically functioning and silent corticotroph adenomas. The analysis of their putative target genes indicates a role of miRNAs in regulation of the corticosteroid receptors expression. Adenomas causing CD have higher expression of hsa-miR-124-3p and hsa-miR-135-5p and lower expression of their target genes NR3C1 and NR3C2. The role of hsa-miR-124-3p in the regulation of NR3C1 was further validated in vitro using AtT-20/D16v-F2 cells. The cells transfected with miR-124-3p mimics showed lower levels of glucocorticoid receptor expression than control cells while the interaction between miR-124-3p and NR3C1 3′ UTR was confirmed using luciferase reporter assay. The results indicate a relatively small difference in miRNA expression between clinically functioning and silent corticotroph pituitary adenomas. High expression of hsa-miR-124-3p in adenomas causing CD plays a role in the regulation of glucocorticoid receptor level and probably in reducing the effect of negative feedback mediated by corticosteroids.
Collapse
|
14
|
Kontogeorgos G, Markussis V, Thodou E, Kyrodimou E, Choreftaki T, Nomikos P, Lampropoulos KI, Tsagarakis S. Association of Pathology Markers with Somatostatin Analogue Responsiveness in Acromegaly. Int J Endocrinol 2022; 2022:8660470. [PMID: 36199813 PMCID: PMC9529452 DOI: 10.1155/2022/8660470] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Somatotroph adenomas (SAs) exhibit a variable responsiveness to somatostatin analogue (SS-a) treatment, a process that is not well understood. We investigated established and novel histological markers as predictors of SS-a responsiveness. METHODS We retrospectively investigated pathology samples from 36 acromegalic patients that underwent transsphenoidal surgery. Clinical, hormonal, and imaging data were available in 24/36 patients, before and after SS-a treatment. Specimens were semiquantitatively analyzed with immunocytochemistry for Ki-67, KER, SSTR-2, SSTR-5, ZAC-1, E-cadherin, and AIP. RESULTS Collectively, 18 (50%) adenomas were each classified as densely/sparsely granulated somatotroph adenomas (DGSAs/SGSAs), respectively. Patients that received preoperative SS-a had lower expression of SSTR-2 compared to those that did not (2.0 (1.0, 3.0) vs. 3.0 (3.0, 3.0), p = 0.042). Compared with DGSAs, SGSAs had higher Ki-67 labeling index (LI) (1.0 (0.5, 1.0) vs. 2.0 (1.0, 3.5), p = 0.013), and a higher proportion of high MR T2 signal (1 (6%) vs. 6 (33%), p = 0.035), and tended to express less ZAC-1 (p = 0.061) and E-cadherin (p = 0.067). In linear regression corrected for baseline growth hormone (GH), ZAC-1 immunostaining was significantly associated with a decrease in GH levels after SS-a treatment (beta (95% confidence interval): -1.53 (-2.80, -0.26), p = 0.021). No markers were associated with changes in circulating insulin-like growth factor-I (IGF-I) after treatment with SS-a. CONCLUSION The novel marker ZAC-1 was associated with GH response to medical treatment with SS-a. The SGSA cases were characterized by higher Ki-67 values and MR T2 signals indicative of an inferior response to SS-a. These findings improve our understanding of the mechanisms underlying SA response to medical treatment.
Collapse
Affiliation(s)
- George Kontogeorgos
- First Propaedeutic Department of Internal Medicine, Division of Endocrinology, 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
| | - Efi Kyrodimou
- Department of Pathology and Pituitary Tumor Reference Center, “G. Gennimatas” General Hospital of Athens, Athens, Greece
| | - Theodossia Choreftaki
- Department of Pathology and Pituitary Tumor Reference Center, “G. Gennimatas” General Hospital of Athens, Athens, Greece
| | | | | | | |
Collapse
|