1
|
Chiloiro S, Giampietro A, Giambò P, Costanza F, Mattogno PP, Lauretti L, Calandrelli R, Gaudino S, Gessi M, Rindi G, Olivi A, De Marinis L, Doglietto F, Bianchi A, Pontecorvi A, Giustina A. IGF-I levels during standard Lanreotide dose predicts biochemical outcome of high-frequency regimen in acromegaly. Pituitary 2024; 28:7. [PMID: 39724447 DOI: 10.1007/s11102-024-01479-9] [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: 11/13/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION First-generation somatostatin receptor ligands (fg-SRLs) are the cornerstone of acromegaly treatment. Additional benefits were shown using high dose (HD) or high frequency (HF), relatively short-term regimens. Although several predictors of response to standard dose (SD)-fg-SRLs were reported, outcome biomarkers for HF administration are not yet available. Here, we aimed to identify predictors of response to long-term HF-fg-SRLs. PATIENTS AND METHODS A retrospective study was performed on 102 patients, treated with Lanreotide. Patients not controlled at 12 months of SD-Lanreotide (120 mg/28 days) were switched to HF-Lanreotide (120 mg/21 days) for additional 12 months. RESULTS Twenty-eight patients were controlled at 6 months of SD-Lanreotide (27.4%); 35 patients were controlled at 12 months of treatment (34.3%). Out of 67 patients treated with HF- Lanreotide, 18 (26.9%) were controlled at 6 months of treatment and remained controlled until 12 months. Both during SD and HF-Lanreotide administrations, IGF-I levels were reduced during the first six months of treatment (p < 0.001), without further significant reduction between 6 and 12 months of therapy. Response at 12 months of SD-Lanreotide was predicted by IGF-I reached at six months of SD-Lanreotide (p = 0.024). Response at 12 months of HF-Lanreotide treatment was predicted by IGF-I levels reached at six months of SD-Lanreotide treatment (p = 0.04) and six months of HF-Lanreotide treatment (p = 0.01). CONCLUSION Our results demonstrated that initial IGF-I levels during SD-Lanreotide predicted the biochemical outcome after 12 months of HF-Lanreotide. Patients in whom HF-Lanreotide did not normalize IGF-I after 6 months of treatment remained uncontrolled 12 months after starting this regimen.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy.
| | - Antonella Giampietro
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Penelope Giambò
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Flavia Costanza
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Pier Paolo Mattogno
- Institute of Neurosurgery, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Liverana Lauretti
- Institute of Neurosurgery, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rosalinda Calandrelli
- ARC Advanced Radiology Center (ARC), Department of Oncological Radiotherapy, and Hematology, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simona Gaudino
- ARC Advanced Radiology Center (ARC), Department of Oncological Radiotherapy, and Hematology, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Gessi
- Anatomic Pathology Unit, Department of Woman and Child Health Sciences and Public Health, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
- Roma-Gemelli European Neuroendocrine Tumor Society Center of Excellence, Rome, Italy
| | - Guido Rindi
- Anatomic Pathology Unit, Department of Woman and Child Health Sciences and Public Health, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
- Roma-Gemelli European Neuroendocrine Tumor Society Center of Excellence, Rome, Italy
| | - Alessandro Olivi
- Institute of Neurosurgery, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura De Marinis
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Francesco Doglietto
- Institute of Neurosurgery, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Antonio Bianchi
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Alfredo Pontecorvi
- Division of Endocrinology and Metabolism, Dipartimento di Medicina e Chirurgia traslazionale, Facoltà di Medicina e Chirurgia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, Number 8, Rome, Italy
| | - Andrea Giustina
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
2
|
Chiloiro S, Moroni R, Giampietro A, Angelini F, Gessi M, Lauretti L, Mattogno PP, Calandrelli R, Tartaglione T, Carlino A, Gaudino S, Olivi A, Rindi G, De Marinis L, Pontecorvi A, Doglietto F, Bianchi A. The Multibiomarker Acro-TIME Score Predicts fg-SRLs Response: Preliminary Results of a Retrospective Acromegaly Cohort. J Clin Endocrinol Metab 2024; 109:1341-1350. [PMID: 37975821 DOI: 10.1210/clinem/dgad673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/15/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
CONTEXT The prompt control of acromegaly is a primary treatment aim for reducing related disease morbidity and mortality. First-generation somatostatin receptor ligands (fg-SRLs) are the cornerstone of medical therapies. A non-negligible number of patients do not respond to this treatment. Several predictors of fg-SRL response were identified, but a comprehensive prognostic model is lacking. OBJECTIVE We aimed to design a prognostic model based on clinical and biochemical parameters, and pathological features, including data on immune tumor microenvironment. METHODS A retrospective, monocenter, cohort study was performed on 67 medically naïve patients with acromegaly. Fifteen clinical, pathological, and radiological features were collected and analyzed as independent risk factors of fg-SRLs response, using univariable and multivariable logistic regression analyses. A stepwise selection method was applied to identify the final regression model. A nomogram was then obtained. RESULTS Thirty-seven patients were fg-SRLs responders. An increased risk to poor response to fg-SRLs were observed in somatotropinomas with absent/cytoplasmatic SSTR2 expression (OR 5.493 95% CI 1.19-25.16, P = .028), with low CD68+/CD8+ ratio (OR 1.162, 95% CI 1.01-1.33, P = .032). Radical surgical resection was associated with a low risk of poor fg-SRLs response (OR 0.106, 95% CI 0.025-0.447 P = .002). The nomogram obtained from the stepwise regression model was based on the CD68+/CD8+ ratio, SSTR2 score, and the persistence of postsurgery residual tumor and was able to predict the response to fg-SRLs with good accuracy (area under the curve 0.85). CONCLUSION Although our predictive model should be validated in prospective studies, our data suggest that this nomogram may represent an easy to use tool for predicting the fg-SRL outcome early.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | | | - Antonella Giampietro
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Flavia Angelini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Marco Gessi
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Liverana Lauretti
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Pier Paolo Mattogno
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Rosalinda Calandrelli
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Tommaso Tartaglione
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Angela Carlino
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Simona Gaudino
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Alessandro Olivi
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Guido Rindi
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Laura De Marinis
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Alfredo Pontecorvi
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Francesco Doglietto
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Antonio Bianchi
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| |
Collapse
|
3
|
Papadimitriou E, Chatzellis E, Dimitriadi A, Kaltsas GA, Theocharis S, Alexandraki KI. Prognostic Biomarkers in Pituitary Tumours: A Systematic Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:42-53. [PMID: 38187082 PMCID: PMC10769480 DOI: 10.17925/ee.2023.19.2.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/07/2023] [Indexed: 01/09/2024]
Abstract
Pituitary tumours (PTs) are the second most common intracranial tumour. Although the majority show benign behaviour, they may exert aggressive behaviour and can be resistant to treatment. The aim of this review is to report the recently identified biomarkers that might have possible prognostic value. Studies evaluating potentially prognostic biomarkers or a therapeutic target in invasive/recurrent PTs compared with either non-invasive or non-recurrent PTs or normal pituitaries are included in this review. In the 28 included studies, more than 911 PTs were evaluated. A systematic search identified the expression of a number of biomarkers that may be positively correlated with disease recurrence or invasion in PT, grouped according to role: (1) insensitivity to anti-growth signals: minichromosome maintenance protein 7; (2) evasion of the immune system: cyclooxygenase 2, arginase 1, programmed cell death protein 1 (PD-1)/programmed death ligand 2, cluster of differentiation (CD) 80/CD86; (3) sustained angiogenesis: endothelial cell-specific molecule, fibroblast growth factor receptor, matrix metalloproteinase 9, pituitary tumour transforming gene; (4) self-sufficiency in growth signals: epidermal growth factor receptor; and (5) tissue invasion: matrix metalloproteinase 9, fascin protein. Biomarkers with a negative correlation with disease recurrence or invasion include: (1) insensitivity to anti-growth signals: transforming growth factor β1, Smad proteins; (2) sustained angiogenesis: tissue inhibitor of metalloproteinase 1; (3) tissue invasion: Wnt inhibitory factor 1; and (4) miscellaneous: co-expression of glial fibrillary acidic protein and cytokeratin, and oestrogen receptors α36 and α66. PD-1/programmed cell death ligand 1 showed no clear association with invasion or recurrence, while cyclin A, cytotoxic T lymphocyte-associated protein 4, S100 protein, ephrin receptor, galectin-3 , neural cell adhesion molecule, protein tyrosine phosphatase 4A3 and steroidogenic factor 1 had no association with invasion or recurrence of PT. With the aim to develop a more personalized approach to the treatment of PT, and because of the limited number of molecular targets currently studied in the context of recurrent PT and invasion, a better understanding of the most relevant of these biomarkers by well-d esigned interventional studies will lead to a better understanding of the molecular profile of PT. This should also meet the increased need of treatable molecular targets.
Collapse
Affiliation(s)
- Eirini Papadimitriou
- First Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Chatzellis
- Endocrinology Diabetes and Metabolism Department, 251 Hellenic Air Force and VA General Hospital, Athens, Greece
| | | | - Gregory A Kaltsas
- First Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
4
|
Osamura RY, Inomoto C, Tahara S, Oyama KI, Matsuno A, Teramoto A. Pathology of Crooke Cells in the Human Pituitaries: A Timely Review. Appl Immunohistochem Mol Morphol 2023; 31:485-489. [PMID: 36251979 DOI: 10.1097/pai.0000000000001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/09/2022] [Indexed: 11/02/2022]
Abstract
Crooke cell change was first found in the regressed and suppressed corticotroph (adrenocorticotropic hormone-producing) cells, and now is known to occur in pituitary tumors. The tumor cells of this type can be recognized by morphology with immunohistochemistry, and are well known to predict aggressive behavior such as invasion and rare metastases. This is one of the representative neuroendocrine tumors in the pituitary which is now considered to have malignant potential as proposed in the pancreas and gastrointestinal tracts. It is important to emphasize the pituitary tumor pathology such as Crooke cell change for prognostication and appropriate therapies. This review article describes the evolution from the Crooke cells to Crooke cell tumors which is timely along with the Fifth WHO classification 2022 published online.
Collapse
Affiliation(s)
- Robert Y Osamura
- Nippon Koukan Hospital, Kawasaki
- Keio University School of Medicine
| | | | | | | | | | | |
Collapse
|
5
|
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
|
6
|
Sabahi M, Ghasemi-Nesari P, Maroufi SF, Shahbazi T, Yousefi O, Shahtaheri SA, Bin-Alamer O, Dabecco R, Velasquez N, Arce KM, Adada B, Benjamin CG, Borghei-Razavi H. Recurrent Cushing Disease: An Extensive Review on Pros and Cons of Different Therapeutic Approaches. World Neurosurg 2023; 172:49-65. [PMID: 36739900 DOI: 10.1016/j.wneu.2023.01.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Recurrent Cushing disease (CD) is characterized by the reappearance of clinical and hormonal aspects of hypercortisolism that occur more than 6 months after an initial post-treatment remission. METHODS We performed a systematic review and meta-analysis to synthesize the evidence about remission and complication rates after transsphenoidal surgery (TSS) radiotherapy (RT) and medical therapy (MT) in recurrent CD patients. A quantitative systematic review was performed. Article selection was performed by searching MEDLINE (using PubMed), and Cochrane electronic bibliographic databases through 2020. RESULTS We noted 61 articles described therapeutic management of recurrent CD patients with representative outcome. A total of 723 patients received different therapeutic modality for their recurrent CD. The remission rates were 0.65 (95% confidence interval [CI] 0.60-0.70), 0.57 (95% CI 0.51-0.63), and 0.75 (95% CI 0.60-0.86) in the TSS, RT, and MT subgroups, respectively. The total remission rate after therapeutic approaches on recurrent CD patients was 0.64 (95% CI 0.60-0.68). A test for subgroup differences revealed there was a statistically significant difference between different subgroups (P = 0.01). The post hoc test showed that in comparison with RT, TSS (P = 0.0344) and MT (P = 0.0149) had a higher rate of remission. However, there was no statistically significant difference between separate therapeutic modalities in terms of complications including diabetes insipidus (P = 1.0) and hypopituitarism (P = 0.28). CONCLUSIONS Compared MT and TSS, RT has a statistically lower rate of remission. Although there is robust superiority of surgery over RT, interpretation of MT data must considered with caution due to the small number of included cases and wide CI range.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Paniz Ghasemi-Nesari
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Farzad Maroufi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Taha Shahbazi
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Yousefi
- Trauma Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Alireza Shahtaheri
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Othman Bin-Alamer
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Rocco Dabecco
- Department of Neurological Surgery, Pauline Braathen Neurological Centre, Cleveland Clinic Florida, Weston, Florida, USA
| | - Nathalia Velasquez
- Department of Otolaryngology/Head and Neck Surgery, Cleveland Clinic Florida, Weston, Florida, USA
| | - Karla M Arce
- Department of Endocrinology, Diabetes and Metabolism, Cleveland Clinic Florida, Weston, Florida, USA
| | - Badih Adada
- Department of Neurological Surgery, Pauline Braathen Neurological Centre, Cleveland Clinic Florida, Weston, Florida, USA
| | | | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Centre, Cleveland Clinic Florida, Weston, Florida, USA.
| |
Collapse
|
7
|
Su D. MCM7 affects the cisplatin resistance of liver cancer cells and the development of liver cancer by regulating the PI3K/Akt signaling pathway. Immunopharmacol Immunotoxicol 2021; 44:17-27. [PMID: 34821526 DOI: 10.1080/08923973.2021.1991372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Aberrant DNA replication is regarded as a component of cancer development. Minichromosome maintenance protein 7 (MCM7), which is critical for the initiation of DNA replication, is overexpressed in multiple malignancies. The effect of MCM7 on cell proliferation, apoptosis, and drug resistance of liver cancer and its mechanism were investigated in this study. METHODS MCM7 expression in normal liver cells, liver cancer cell lines, and tissues, as well as adjacent tissues, was determined by qRT-PCR. CCK-8 and flow cytometry was performed to detect cell viability, apoptosis, and cell cycle, respectively. The related mRNA and protein expressions were detected by qRT-PCR and western blot. RESULTS High expression of MCM7 was found in liver cancer tissues and cells, which results in notably lower survival time of patients. Cisplatin (DDP) could inhibit cell proliferation and affect MCM7 expression. Silencing of MCM7 inhibited cell viability, promoted cell apoptosis, arrested cell cycle at G1 phase, and enhanced the effect of DDP on cancer cells, while overexpression of MCM7 did the opposite. Moreover, silencing of MCM7 inhibited cyclinD1 and Ki-67 expressions. The overexpression of MCM7 increased phosphorylation levels of PI3K and AKT, activated the PI3K/AKT pathway, and weakened the inhibitory effect of DDP on the PI3K/AKT pathway. CONCLUSION Silencing of MCM7 may inhibit cell proliferation and promote apoptosis by regulating the PI3K/AKT pathway to affect the cell cycle, thus affecting the development of liver cancer, and improving the sensitivity of liver cancer cells to DDP.
Collapse
Affiliation(s)
- Dongna Su
- Department of Infectious Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
8
|
Tang Y, Xie T, Wu S, Yang Q, Liu T, Li C, Liu S, Shao Z, Zhang X. Quantitative proteomics revealed the molecular characteristics of distinct types of granulated somatotroph adenomas. Endocrine 2021; 74:375-386. [PMID: 34043183 DOI: 10.1007/s12020-021-02767-1] [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] [Received: 12/24/2020] [Accepted: 05/15/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Somatotroph adenomas are obviously heterogeneous in clinical characteristics, imaging performance, pathological diagnosis and therapeutic effect. The heterogeneity of the tumors, especially for SG and DG type adenomas, have attracted great interest in identifying the specific pathological markers and therapeutic targets of them. However, previous analyses of the molecular characteristics of the subtypes of somatotroph adenomas were performed at genomic and transcriptome level. The proteomic differences between the two subtypes of somatotroph adenomas are still unknown. METHODS Tumor samples were surgically removed from 10 sporadic pituitary somatotroph adenoma patients and grouped according to the pathological type. Tandem mass tag (TMT)-based quantitative proteomic analysis was employed to analyze the proteomic differences between SG and DG tumors. RESULTS In total, 228 differentially expressed proteins were identified between SG adenomas and DG adenomas. They were enriched mainly in extracellular matrix (ECM)-receptor interaction, leukocyte transendothelial migration, arrhythmogenic right ventricular cardiomyopathy and DNA replication pathways. Protein-protein interaction (PPI) network analysis indicated that Cadherin-1 and Catenin beta-1 were the most important key proteins in the differences between SG and DG adenomas. Immunohistochemistry (IHC) confirmed the expression levels of the key proteins. CONCLUSIONS This study provides large-scale proteome molecular characteristics of distinct granulation subtypes of somatotroph adenomas. Compared with DG adenomas, The differential protein of SG adenomas mostly enrich in invasive and proliferative functions and pathways at the proteomic level. Cadherin-1 and Catenin beta-1 play key roles in the different biological characteristics of the two tumor subtypes.
Collapse
Affiliation(s)
- Yifan Tang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Silin Wu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiaoqiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tengfei Liu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Li
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuang Liu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiyong Shao
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Digital Medical Research Center, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Medical Image Computing and Computer-Assisted Intervention, Shanghai, China.
| |
Collapse
|
9
|
Mattogno PP, D’Alessandris QG, Chiloiro S, Bianchi A, Giampietro A, Pontecorvi A, De Marinis L, Olivi A, Anile C, Lauretti L. Reappraising the Role of Trans-Sphenoidal Surgery in Prolactin-Secreting Pituitary Tumors. Cancers (Basel) 2021; 13:cancers13133252. [PMID: 34209686 PMCID: PMC8269319 DOI: 10.3390/cancers13133252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/13/2021] [Accepted: 06/24/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Prolactinomas constitute a subgroup of pituitary adenomas for which there are several treatment options. Dopamine agonists (DA), since their introduction, have shown a strong efficacy both in the control of hyperprolactinemia and of the significant volumetric reduction of prolactinomas, leading, in some cases, to a definitive cure. Trans-sphenoidal surgery (TSS) has been traditionally confined to a failure of medical therapy, pituitary apoplexy with neurological worsening, and prolactinomas with wide cystic components. Moreover, the recent technical innovations introduced in TSS and increasing experience of surgeons have allowed to achieve better results, such as complete tumor resection with lower complication rates. On these grounds, the authors reviewed the extensive institutional Prolactinomas case series over the last 25 years to analyze the role of TSS in the management of Prolactinomas, particularly in terms of the cure rate. Abstract Background: Prolactinomas represent a unique challenge for endocrinologists and neurosurgeons. Considering recent innovations in surgical practice, the authors aimed to investigate the best management for prolactinomas. Methods: A retrospective, cross-sectional and monocentric study was designed. Consecutive patients affected by prolactinomas were enrolled if treated with a first-line treatment with a dopamine agonist (DA) or trans-sphenoidal surgery (TSS). Patients carried giant prolactinomas, and those with a follow-up <12 months were excluded. Results: Two hundred and fifty-nine patients were enrolled. The first treatment was DA for 140 patients and TS for 119 cases. One hundred and forty-six of 249 patients (58.6%) needed a second therapy. The mean follow-up was 102.2 months (12–438 months). Surgery highly impacted on the cure rate—in particular, in females (p = 0.0021) and in microprolactinomas (p = 0.0020). Considering the multivariate analysis, the female gender and surgical treatment in the course of the clinical history were the only independent positive predictors of a cure at the end of 5 years follow-up (p = 0.0016, p = 0.0005). The evaluation of serum prolactin (24 hours after TSS) revealed that 86.4% of patients with postoperative prolactin (PRL) ≤10 ng/mL were cured at the end of the follow-up (p < 0.0001). Conclusions: According to our experience, surgery allows a high cure rate of prolactinomas, particularly in females with microadenoma, with a good safety profile. TSS for prolactinomas should be considered as a concrete option, during the multidisciplinary evaluation, in centers of reference for pituitary diseases.
Collapse
Affiliation(s)
- Pier Paolo Mattogno
- Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (Q.G.D.); (A.O.); (C.A.); (L.L.)
- Correspondence:
| | - Quintino Giorgio D’Alessandris
- Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (Q.G.D.); (A.O.); (C.A.); (L.L.)
| | - Sabrina Chiloiro
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Gemelli IRCCS-Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (S.C.); (A.B.); (A.G.); (A.P.); (L.D.M.)
| | - Antonio Bianchi
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Gemelli IRCCS-Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (S.C.); (A.B.); (A.G.); (A.P.); (L.D.M.)
| | - Antonella Giampietro
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Gemelli IRCCS-Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (S.C.); (A.B.); (A.G.); (A.P.); (L.D.M.)
| | - Alfredo Pontecorvi
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Gemelli IRCCS-Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (S.C.); (A.B.); (A.G.); (A.P.); (L.D.M.)
| | - Laura De Marinis
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Gemelli IRCCS-Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (S.C.); (A.B.); (A.G.); (A.P.); (L.D.M.)
| | - Alessandro Olivi
- Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (Q.G.D.); (A.O.); (C.A.); (L.L.)
| | - Carmelo Anile
- Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (Q.G.D.); (A.O.); (C.A.); (L.L.)
| | - Liverana Lauretti
- Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore-Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (Q.G.D.); (A.O.); (C.A.); (L.L.)
| |
Collapse
|
10
|
[The 2017 WHO classification of pituitary tumors]. DER PATHOLOGE 2021; 42:333-351. [PMID: 33877399 DOI: 10.1007/s00292-021-00932-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 10/21/2022]
Abstract
The 2017 WHO classification of pituitary tumors is still based on structural analyses and expression of various pituitary hormones. Three innovations have to be considered: (1) The expression of pituitary transcription factors Pit‑1, T‑Pit and SF‑1. (2) The term "atypical adenoma" was replaced by "aggressive adenoma". (3) The three tumor types of the neurohypophysis (pituicytoma, spindle cell oncocytoma, granular cell tumor) are defined by their common expression of TTF‑1. Craniophyryngiomas are identified as adamantinomatous type by focal nuclear expression of β‑catenin or as papillary type by demonstration of BRAF V600E mutation. Further primary tumors of the pituitary are extremely rare. These and also the other tumors of the sellar region can be structurally very similar to pituitary adenomas but can be-nearly without exception-differentiated by immunocytochemistry.
Collapse
|
11
|
Yamamoto M, Nakao T, Ogawa W, Fukuoka H. Aggressive Cushing's Disease: Molecular Pathology and Its Therapeutic Approach. Front Endocrinol (Lausanne) 2021; 12:650791. [PMID: 34220707 PMCID: PMC8242934 DOI: 10.3389/fendo.2021.650791] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Cushing's disease is a syndromic pathological condition caused by adrenocorticotropic hormone (ACTH)-secreting pituitary adenomas (ACTHomas) mediated by hypercortisolemia. It may have a severe clinical course, including infection, psychiatric disorders, hypercoagulability, and metabolic abnormalities, despite the generally small, nonaggressive nature of the tumors. Up to 20% of ACTHomas show aggressive behavior, which is related to poor surgical outcomes, postsurgical recurrence, serious clinical course, and high mortality. Although several gene variants have been identified in both germline and somatic changes in Cushing's disease, the pathophysiology of aggressive ACTHomas is poorly understood. In this review, we focused on the aggressiveness of ACTHomas, its pathology, the current status of medical therapy, and future prospects. Crooke's cell adenoma (CCA), Nelson syndrome, and corticotroph pituitary carcinoma are representative refractory pituitary tumors that secrete superphysiological ACTH. Although clinically asymptomatic, silent corticotroph adenoma is an aggressive ACTH-producing pituitary adenoma. In this review, we summarize the current understanding of the pathophysiology of aggressive ACTHomas, including these tumors, from a molecular point of view based on genetic, pathological, and experimental evidence. The treatment of aggressive ACTHomas is clinically challenging and usually resistant to standard treatment, including surgery, radiotherapy, and established medical therapy (e.g., pasireotide and cabergoline). Temozolomide is the most prescribed pharmaceutical treatment for these tumors. Reports have shown that several treatments for patients with refractory ACTHomas include chemotherapy, such as cyclohexyl-chloroethyl-nitrosourea combined with 5-fluorouracil, or targeted therapies against several molecules including vascular endothelial growth factor receptor, cytotoxic T lymphocyte antigen 4, programmed cell death protein 1 (PD-1), and ligand for PD-1. Genetic and experimental evidence indicates that some possible therapeutic candidates are expected, such as epidermal growth factor receptor tyrosine kinase inhibitor, cyclin-dependent kinase inhibitor, and BRAF inhibitor. The development of novel treatment options for aggressive ACTHomas is an emerging task.
Collapse
Affiliation(s)
- Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe, Japan
| | | | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Kobe University Hospital, Kobe, Japan
- *Correspondence: Hidenori Fukuoka,
| |
Collapse
|
12
|
Chiloiro S, Bima C, Tartaglione T, Giampietro A, Gessi M, Lauretti L, Anile C, Colosimo C, Rindi G, Pontecorvi A, De Marinis L, Bianchi A. Pasireotide and Pegvisomant Combination Treatment in Acromegaly Resistant to Second-Line Therapies: A Longitudinal Study. J Clin Endocrinol Metab 2019; 104:5478-5482. [PMID: 31219586 DOI: 10.1210/jc.2019-00825] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/13/2019] [Indexed: 12/27/2022]
Abstract
CONTEXT The treatment of acromegaly resistant to first- and second-line therapies can be extremely challenging. DESIGN We have described six patients who were successfully treated with a combination therapy of pasireotide and pegvisomant and compared them with a control group of patients resistant to conventional somatostatin analogs (SSAs), whose disease was controlled with other treatment, such as pasireotide (as monotherapy) or pegvisomant (as monotherapy or combined with conventional SSAs). RESULTS In these six patients, acromegaly was controlled with combined pasireotide and pegvisomant treatment after failure of all other treatments. Compared with the 49 patients in the control group, these six patients had giant and invasive pituitary adenomas (at both the cavernous sinus and other structures). Although not statistically significant, higher growth hormone levels, more elevated Ki-67 expression, greater somatostatin receptor (SSTR) subtype 5 expression, and lower SSTR subtype 2 expression at the diagnosis of acromegaly were detected in patients receiving combination treatment with pasireotide and pegvisomant compared with the control group. CONCLUSION Our data have reinforced the importance of personalized treatment of patients with acromegaly according to the clinical, biochemical, molecular, and morphological disease markers and suggest that combined treatment with pasireotide and pegvisomant can induce disease control in tumors with low SSTR2 expression, resistant to conventional SSAs (alone or combined with pegvisomant) and to new-generation SSAs alone (pasireotide).
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara Bima
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tommaso Tartaglione
- UOC di Radiologia e Diagnostica per Immagini, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Giampietro
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Gessi
- Dipartimento di Anatomia Patologica, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Liverana Lauretti
- Dipartimento di Neurochirurgia, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Carmelo Anile
- Dipartimento di Neurochirurgia, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Cesare Colosimo
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Guido Rindi
- Dipartimento di Anatomia Patologica, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
- Istituto di Patologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alfredo Pontecorvi
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura De Marinis
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Bianchi
- Dipartimento di Endocrinologia, Istituto di Patologia Speciale Medica, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
13
|
Ban X, Yan J, Yu S, Lu Z, Chang X, Jia C, Gao C, Shao H, Wu Y, Mao X, Zhang Y, Li Y, Chen J. High minichromosome maintenance protein 7 proliferation indices: a powerful predictor of progression in pancreatic neuroendocrine neoplasms without distant metastasis at the time of surgery. Hum Pathol 2018; 85:101-111. [PMID: 30447299 DOI: 10.1016/j.humpath.2018.10.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 12/28/2022]
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) have an unpredictable clinical course that varies from indolent to highly malignant. No immunohistochemical markers are available for reliable prediction of the biological behavior of early stage PanNENs. Minichromosome maintenance protein 7 (MCM7) is a putative powerful marker of cell proliferation. Whether the expression of MCM7 is related to the risk of PanNENs progression remains unclear. We assessed the clinical behavior of 156 PanNENs with respect to stage, grade, Ki-67 index, MCM7 index, and other pathologic features. A high MCM7 index was significantly associated with larger tumor size (P < .001), nonfunctioning tumor (P < .001), increased grade (P < .0001), and later TNM stage (P < .001). In multivariate analysis, G2/G3 (hazard ratio [HR], 2.21; 95% confidence interval [CI], 1.35-3.62; P < .001), stage III/IV (HR, 2.11; 95% CI, 1.31-3.41; P < .001), and MCM7 labeling index >5% (HR, 3.81; 95% CI, 1.30-11.17; P = .02) were independent negative prognostic factors related to the risk of tumor progression in stage I-IV disease. MCM7 labeling index >5% was associated with an increased risk of progression in stages I-V, I-III, and I-II. Our study confirms that MCM7 is a valuable marker for assessing the progression of PanNENs, especially in patients with early stage disease and without distant metastasis.
Collapse
Affiliation(s)
- Xinchao Ban
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Jie Yan
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Shuangni Yu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Zhaohui Lu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Cen Gao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Huilin Shao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Yan Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Xinxin Mao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Yue Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Yuan Li
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences-Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
14
|
Liao X, Han C, Wang X, Huang K, Yu T, Yang C, Huang R, Liu Z, Han Q, Peng T. Prognostic value of minichromosome maintenance mRNA expression in early-stage pancreatic ductal adenocarcinoma patients after pancreaticoduodenectomy. Cancer Manag Res 2018; 10:3255-3271. [PMID: 30233242 PMCID: PMC6130532 DOI: 10.2147/cmar.s171293] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background The aim of the current study was to investigate the potential prognostic value of minichromosome maintenance (MCM) genes in patients with early-stage pancreatic ductal adenocarcinoma (PDAC) after pancreaticoduodenectomy by using the RNA-sequencing dataset from The Cancer Genome Atlas (TCGA). Methods An RNA-sequencing dataset of 112 early-stage PDAC patients who received a pancreaticoduodenectomy was obtained from TCGA. Survival analysis was used to identify potential prognostic values of MCM genes in PDAC overall survival (OS). Results Through mining public databases, we observed that MCM genes (MCM2, MCM3, MCM4, MCM5, MCM6, and MCM7) were upregulated in pancreatic cancer tumor tissue and have a strong positive coexpression with each other. Multivariate survival analysis indicated that a high expression of MCM4 significantly increased the risk of death in patients with PDAC, and time-dependent receiver operating characteristic analysis showed an area under the curve of 0.655, 0.587, and 0.509 for a 1-, 2-, and 3-year PDAC OS prediction, respectively. Comprehensive survival analysis of MCM4 using stratified and joint effects survival analysis suggests that MCM4 may be an independent prognostic indicator for PDAC OS. Gene set enrichment analysis indicated that MCM4 may participate in multiple biologic processes and pathways, including DNA replication, cell cycle, tumor protein p53, and Notch signaling pathways, thereby affecting prognosis of PDAC patients. Conclusions Our study indicates that MCM2–7 were upregulated in pancreatic cancer tumor tissues, and mRNA expression of MCM4 may serve as an independent prognostic indicator for PDAC OS prediction after pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China,
| |
Collapse
|
15
|
Todnem N, Ward A, Segar S, Rojiani AM, Rahimi SY. Clinically Silent Adrenocorticotropic Hormone-Positive Crooke Cell Adenoma: Case Report and Review of Literature. World Neurosurg 2018; 119:197-200. [PMID: 30077745 DOI: 10.1016/j.wneu.2018.07.233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Pituitary adenomas are one of the most common tumors of adulthood; however, subtypes such as Crooke cell adenoma are relatively rare. CASE DESCRIPTION We present the case of a 55-year-old woman who presented with new-onset intermittent headache and dizziness. Clinical and laboratory investigations were not suggestive of corticotroph tumor. However, subsequent computed tomography and magnetic resonance imaging scans revealed the presence of a suprasellar pituitary adenoma displacing the optic chiasma superiorly, with hemorrhage and sellar expansion. The lesion was removed by transsphenoidal surgery and the biopsy confirmed the lesion to be a nonfunctioning pituitary macroadenoma. Further investigation revealed that the specimen demonstrated Crooke hyaline changes, with strong immunoreactivity for adrenocorticotropic hormone. However, initial workup and postoperative testing lacked evidence of Cushing disease. There was no sign of recurrence after 1-year follow-up. CONCLUSIONS Clinically silent Crooke cell adenomas are rare occurrences, and as such we report this case with investigation of past cases.
Collapse
Affiliation(s)
- Nathan Todnem
- Department of Neurosurgery, The Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ayobami Ward
- Department of Neurosurgery, The Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Sharmila Segar
- Department of Neurosurgery, The Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Amyn M Rojiani
- Department of Pathology, The Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Scott Y Rahimi
- Department of Neurosurgery, The Medical College of Georgia at Augusta University, Augusta, Georgia, USA.
| |
Collapse
|
16
|
Yan J, Du P, Jia Y, Chang Z, Gan S, Xu X, Wang Y, Qin Y, Kan Q. Ablation of MCM10 using CRISPR/Cas9 restrains the growth and migration of esophageal squamous cell carcinoma cells through inhibition of Akt signaling. Onco Targets Ther 2018; 11:3323-3333. [PMID: 29922071 PMCID: PMC5995424 DOI: 10.2147/ott.s157025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Minichromosome maintenance 10 (MCM10) is deregulated in several malignancies including cervical cancer and urothelial carcinoma. However, the expression and biologic role of MCM10 in esophageal squamous cell carcinoma (ESCC) is still unknown. Methods In this study, we performed immunohistochemistry and real-time polymerase chain reaction (PCR) analysis to examine the expression of MCM10 in ESCC and adjacent normal esophageal tissues. The associations of MCM10 expression with clinicopathologic parameters of ESCC were analyzed. Ablation of MCM10 through the CRISPR/Cas9 technology was conducted and its impact on ESCC cell growth and migration was investigated. Results The mRNA and protein expression levels of MCM10 were significantly greater in ESCC than in normal tissues (P<0.001). The expression of MCM10 was significantly associated with age at diagnosis (P=0.033), but not with gender, differentiation grade, invasion status, or tumor–node–metastasis (TNM) stage. Knockout of MCM10 significantly suppressed the proliferation, colony formation, and migration capacity of EC109 ESCC cells, compared to control cells harboring wild-type MCM10. Mechanistically, MCM10 depletion markedly reduced the phosphorylation of Akt. Overexpression of constitutively active Akt significantly restored the aggressive phenotype of MCM10-null EC109 cells. Conclusion In conclusion, these results suggest that MCM10 acts as an oncogene in ESCC through activation of Akt signaling and represents a promising therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Jie Yan
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pan Du
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, Zhengzhou, China
| | - Yongxu Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Silin Gan
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohan Xu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Quancheng Kan
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Liao X, Liu X, Yang C, Wang X, Yu T, Han C, Huang K, Zhu G, Su H, Qin W, Huang R, Yu L, Deng J, Zeng X, Ye X, Peng T. Distinct Diagnostic and Prognostic Values of Minichromosome Maintenance Gene Expression in Patients with Hepatocellular Carcinoma. J Cancer 2018; 9:2357-2373. [PMID: 30026832 PMCID: PMC6036720 DOI: 10.7150/jca.25221] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/14/2018] [Indexed: 12/31/2022] Open
Abstract
Background: The aim of the present study was to identify diagnostic and prognostic values of minichromosome maintenance (MCM) gene expression in patients with hepatocellular carcinoma (HCC). Methods: The biological function of the MCM genes were investigated by bioinformatics analysis. The diagnostic and prognostic values of the MCM genes were investigated by using the data of HCC patients from the GSE14520 and The Cancer Genome Atlas (TCGA) databases. Results: Bioinformatics analysis of the MCM genes substantiated that MCM2-7 genes were significantly enriched in DNA replication and cell cycle, and co-expressed with each other. These genes also co-expressed in HCC tumor tissue in both the GSE14520 and TCGA cohort. We also observed that the expression of the MCM2-7 genes was increased in tumor tissue, and diagnostic receiver operating characteristic analysis of MCM2-7 indicated that these genes could serve as sensitive diagnostic markers in HCC. Survival analysis in the GSE14520 cohort suggested that expression of MCM2, MCM4, MCM5, and MCM6 were significantly associated with hepatitis B virus-related HCC overall survival (OS). However, none of the MCM genes were associated with recurrence-free survival in the GSE14520 cohort. The validation cohort of TCGA suggested that the expression of MCM2, MCM6, and MCM7 were significantly correlated with HCC OS. Conclusion: Our study indicated that MCM2-7 genes may be potential diagnostic biomarkers in patients with HCC. Among them, MCM2 and MCM6 may serve as potential prognostic biomarkers for HCC.
Collapse
Affiliation(s)
- Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.,Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong Province, People's Republic of China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Long Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jianlong Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.,Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, 537000, Guangxi, China
| | - Xianmin Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| |
Collapse
|
18
|
Neves H, Kwok HF. In sickness and in health: The many roles of the minichromosome maintenance proteins. Biochim Biophys Acta Rev Cancer 2017; 1868:295-308. [DOI: 10.1016/j.bbcan.2017.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/29/2017] [Accepted: 06/01/2017] [Indexed: 01/09/2023]
|
19
|
Garbicz F, Mehlich D, Rak B, Sajjad E, Maksymowicz M, Paskal W, Zieliński G, Włodarski PK. Increased expression of the microRNA 106b~25 cluster and its host gene MCM7 in corticotroph pituitary adenomas is associated with tumor invasion and Crooke's cell morphology. Pituitary 2017; 20:450-463. [PMID: 28432562 PMCID: PMC5508039 DOI: 10.1007/s11102-017-0805-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE MCM7 (minichromosome maintenance complex component 7), a DNA replication licensing factor, is a host gene for the oncogenic miR-106b~25 cluster. It has been recently revealed as a relevant prognostic biomarker in a variety of cancers, including pituitary adenomas. The purpose of this study was to assess whether miR-106b~25 and MCM7 levels correlate with tumor invasiveness in a cohort of ACTH-immunopositive adenomas. METHODS Tissue samples were obtained intraoperatively from 25 patients with pituitary adenoma. Tumor invasiveness was assessed according to the Knosp grading scale. MCM7, Ki-67 and TP53 levels were assessed by immunohistochemical staining, while the expression of miR-106b-5p, miR-93-5p, miR-93-3p and miR-25-3p were measured using quantitative real-time PCR performed on RNA isolated from FFPE tissues. RESULTS We have found a significant increase in MCM7 and Ki-67 labeling indices in invasive ACTHomas. Moreover, MCM7 was ubiquitously overexpressed in Crooke's cell adenomas. The expression of miR-93-5p was significantly elevated in invasive compared to noninvasive tumors. In addition, all four microRNAs from the miR-106b~25 cluster displayed marked upregulation in Crooke's cell adenomas. Remarkably, MCM7 and miR-106b-5p both strongly correlated with Knosp grade. A combination of MCM7 LI and miR-106b~25 cluster expression was able to accurately differentiate invasive from noninvasive tumors and had a significant discriminatory ability to predict postoperative tumor recurrence/progression. CONCLUSIONS miR-106b~25 and its host gene MCM7 are potential novel biomarkers for invasive ACTH-immunopositive pituitary adenomas. Additionally, they are both significantly upregulated in rare Crooke's cell adenomas and might therefore contribute to their aggressive phenotype.
Collapse
Affiliation(s)
- Filip Garbicz
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland
| | - Dawid Mehlich
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland
| | - Beata Rak
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Warsaw, Poland
- Department of Internal Diseases and Endocrinology, Public Central Teaching Hospital Medical University of Warsaw, Warsaw, Poland
| | - Emir Sajjad
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland
- Department of Neurosurgery, Military Institute of Medicine, Warsaw, Poland
| | - Maria Maksymowicz
- Department of Pathology and Laboratory Diagnostics, M. Skłodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Wiktor Paskal
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland
| | - Grzegorz Zieliński
- Department of Neurosurgery, Military Institute of Medicine, Warsaw, Poland
| | - Paweł K Włodarski
- Laboratory of Centre for Preclinical Research, Department of Histology and Embryology, Medical University of Warsaw, Banacha 1B, 02-091, Warsaw, Poland.
| |
Collapse
|
20
|
Qiu YT, Wang WJ, Zhang B, Mei LL, Shi ZZ. MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway. Oncol Rep 2017; 37:3590-3596. [PMID: 28498460 DOI: 10.3892/or.2017.5614] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 04/24/2017] [Indexed: 11/06/2022] Open
Abstract
The roles and mechanisms of mini-chromosome maintenance complex component 7 (MCM7) amplification and overexpression in esophageal carcinogenesis were investigated. By analyzing the TCGA datasets, we found that MCM7 was amplified in approximately 12% of esophageal squamous cell carcinomas (ESCCs), and in more than 4% of head and neck squamous cell carcinomas and stomach carcinomas. Overexpression of MCM7 was further verified in three independent GEO datasets of esophageal cancer. Knockdown of MCM7 using two siRNAs significantly inhibited cell proliferation, colony formation and migration of KYSE510 and EC9706 cells in vitro. Noteworthy, we further found that silencing of MCM7 suppressed the phosphorylation of AKT1 and mTOR both in KYSE510 and EC9706 cells, and reduced the cell cycle regulatory proteins cyclin D1, cyclin E2 and CDK2. Taken together, our findings suggested that MCM7 promoted tumor cell proliferation, colony formation and migration of ESCC cells via activating AKT1/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yun-Tan Qiu
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Wen-Jun Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Bing Zhang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Li-Li Mei
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Zhi-Zhou Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
21
|
Tamilzhalagan S, Rathinam D, Ganesan K. Amplified 7q21-22 geneMCM7and its intronic miR-25 suppressCOL1A2associated genes to sustain intestinal gastric cancer features. Mol Carcinog 2017; 56:1590-1602. [DOI: 10.1002/mc.22614] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 12/09/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Sembulingam Tamilzhalagan
- Unit of Excellence in Cancer Genetics; Department of Genetics; Centre for Excellence in Genomic Sciences; School of Biological Sciences; Madurai Kamaraj University; Madurai India
| | - Dhanasekaran Rathinam
- Unit of Excellence in Cancer Genetics; Department of Genetics; Centre for Excellence in Genomic Sciences; School of Biological Sciences; Madurai Kamaraj University; Madurai India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics; Department of Genetics; Centre for Excellence in Genomic Sciences; School of Biological Sciences; Madurai Kamaraj University; Madurai India
| |
Collapse
|