1
|
Carmona-Bayonas A, Álvarez Escola C, Ballester Navarro I, Hernando Cubero J, Mangas Cruz MÁ, Garcia-Centeno R, Iglesias C, García-Donas J, Picón MJ, Paja M, González Batanero L, García L, Alonso Gordoa T, López C, Hanzu F, Martínez Trufero J, Febrero Sánchez B, Saiz López P, Blanco Carrera C, Ramón Y Cajal T, Veiguela B, Gressani O, Valdés N, Jimenez-Fonseca P. Does adjuvant mitotane impact cure rates in adrenocortical carcinoma? Insights from the ICARO-GETTHI/SEEN registry. J Clin Endocrinol Metab 2025:dgaf082. [PMID: 39988973 DOI: 10.1210/clinem/dgaf082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy with a high risk of postoperative recurrence. Although adjuvant mitotane is commonly used, its potential for achieving cure rather than simply postponing inevitable recurrence remains uncertain. This study investigates whether mitotane impacts ACC recurrence patterns by preventing or delaying recurrence. METHODS This retrospective analysis used data from the ICARO-GETTHI/SEEN registry, which includes 36 Spanish hospitals. Recurrence in non-metastatic ACC patients after resection was analyzed using Cox models, flexible longitudinal models, and mixture cure models to evaluate the impact of mitotane. RESULTS Among 244 patients, 133 (52%) received adjuvant mitotane, with therapeutic levels monitored in 84%. Findings suggest a possible "cure fraction" with a 32.5% estimated 30-year cure rate (95% CI, 23.4%-45.0%). Cox regression indicated a 39% reduced recurrence risk (HR 0.61; 95% CI, 0.39-0.95) for mitotane-treated patients, with effects diminishing over 24 months. Mixture cure models suggest mitotane primarily delays rather than prevents recurrence. Effect modification analysis showed significant benefit in males (HR 0.33; 95% CI, 0.16-0.69), younger patients, tumors with higher Ki-67% (modeled as a continuous variable), and those with venous invasion (HR 0.47; 95% CI, 0.27-0.82), with potential synergy when combined with radiotherapy. CONCLUSION This study underscores the intriguing possibility that adjuvant mitotane delays recurrence, yet questions remain as to its curative capacity. The early benefit suggests a cytostatic effect, but certain subgroups-especially males, younger individuals, and those with high-risk tumors-may experience a more durable outcome. Further research is needed to explore mitotane's curative potential in ACC management.
Collapse
Affiliation(s)
- Alberto Carmona-Bayonas
- Medical Oncology Department. Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria (IMIB), Universidad de Murcia (UMU), Murcia, Spain
| | | | | | - Jorge Hernando Cubero
- Gastrointestinal and Endocrine Tumors Unit, Medical Oncology Department. Hospital Universitario Val de Hebrón, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Rogelio Garcia-Centeno
- Endocrinology and Nutrition Department. Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - Clara Iglesias
- Medical Oncology Department. Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Jesús García-Donas
- Medical Oncology Department. Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - María José Picón
- Endocrinology and Nutrition Department. Hospital Universitario Virgen de La Victoria, Biomedical Research Institute-IBIMA, Málaga, CIBER Pathophysiology of Obesity and Nutrition-CIBERON, Madrid, Spain
| | - Miguel Paja
- Endocrinology Department. Hospital Universitario de Basurto, Bilbao, University of the Basque Country, University of Pais Vasco (UPV/EHU), Spain
| | | | - Lourdes García
- Endocrinology and Nutrition Department. Hospital Universitario de Jerez, Jerez, Spain
| | - Teresa Alonso Gordoa
- Medical Oncology Department. Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Carlos López
- Medical Oncology Department. Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Valdecilla (IDIVAL), Universidad de Cantabria (UNICAN), Santander, Spain
| | - Felicia Hanzu
- Endocrinology and Nutrition Department. Hospital Clinic, Barcelona, Spain
| | | | - Beatriz Febrero Sánchez
- Endocrine Surgery Unit, General Surgery Department. Hospital Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Patricia Saiz López
- Medical Oncology Department. Hospital Universitario de Burgos, Burgos, Spain
| | - Concepción Blanco Carrera
- Endocrinology and Nutrition Department. Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Teresa Ramón Y Cajal
- Medical Oncology Department. Hospital de la Santa Creu y Sant Pau, Barcelona, Spain
| | - Brenda Veiguela
- Endocrinology and Nutrition Department. Hospital Universitario de Cabueñes, Gijón, Spain
| | - Oswaldo Gressani
- Interuniversity Institute for Biostatistics and statistical Bioinformatics (I-BioStat), Data Science Institute, Hasselt University, Hasselt, Belgium
| | - Nuria Valdés
- Endocrinology and Nutrition Department. Hospital Universitario de Cruces, Biobizkaia, University of Pais Vasco (UPV/EHU), CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Paula Jimenez-Fonseca
- Medical Oncology Department. Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| |
Collapse
|
2
|
Ma S, Wu L, Ye L, Habra MA, Balderrama-Brondani V, Wang W. Adjuvant radiation therapy improves outcome of patients with surgical resected adrenocortical carcinoma. Endocrine 2025:10.1007/s12020-025-04163-5. [PMID: 39862362 DOI: 10.1007/s12020-025-04163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE Adrenocortical carcinoma (ACC) is a rare malignancy known for high rates of recurrence and poor prognosis. Previous studies revealed controversial roles of adjuvant radiation therapy (RT) in patient management. This study aimed to investigate the role of adjuvant RT in postoperative ACC patients. METHODS Patients with histologically confirmed ACC who underwent surgical resection in the Surveillance, Epidemiology, and End Results (SEER) database between 2004 and 2020 were enrolled. Propensity score matching (PSM) was used to balance baseline characteristics between patients receiving adjuvant RT and patients who did not receive RT. Overall survival (OS) and recurrence-free survival (RFS) was analyzed using the Kaplan-Meir method. Risk factors associated with survival outcome was analyzed by univariate and multivariate Cox regression analyses. Subgroup analyses were stratified by European Network for the Study of Adrenal Tumors (ENSAT) disease stage or chemotherapy. A joint retrospective analysis of stage III patients was performed based on data obtained from Ruijin Hospital and MD Anderson Cancer Center. RESULTS Among the 700 patients enrolled, 137 patients undergoing postoperative RT were matched with 137 patients who did not receive RT. Overall survival for patients with adjuvant RT was better than patients without RT (log-rank P = 0.015). The 3-year and 5-year OS were 55.2 and 47.1% for patients with RT, vs 42.6 and 34.0% for patients without RT. Multivariate analysis showed adjuvant RT was independently associated with lower mortality (hazard ratio [HR] 0.63, P = 0.007). Subgroup analysis stratified by disease stage demonstrated that adjuvant RT showed the most favorable effect in stage III patients (HR 0.53, P = 0.013). Furthermore, joint analysis of two centers showed a tendency of better OS and local control rate for stage III patients with mitotane plus RT than those with mitotane alone. CONCLUSION Our study indicated that adjuvant RT was associated with improved prognosis for ACC patients, especially for patients with ENSAT stage III diseases. Integrating adjuvant RT into standard care of ACC may be considered.
Collapse
Affiliation(s)
- Shuqing Ma
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Luming Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mouhammed Amir Habra
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vania Balderrama-Brondani
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Tsuboi I, Kardoust Parizi M, Matsukawa A, Mancon S, Miszczyk M, Schulz RJ, Fazekas T, Cadenar A, Laukhtina E, Kawada T, Katayama S, Iwata T, Bekku K, Wada K, Remzi M, Karakiewicz PI, Araki M, Shariat SF. The efficacy of adjuvant mitotane therapy and radiotherapy following adrenalectomy in patients with adrenocortical carcinoma: A systematic review and meta-analysis. Urol Oncol 2024:S1078-1439(24)00654-9. [PMID: 39379210 DOI: 10.1016/j.urolonc.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare and aggressive malignancy with a high recurrence rate after surgical therapy with curative intent. Adjuvant radiotherapy (RT) and mitotane therapy have been proposed as options following the adrenalectomy. However, the efficacy of adjuvant RT or mitotane therapy remains controversial. We aimed to evaluate the efficacy of adjuvant therapy in patients who underwent adrenalectomy for localised ACC. The PubMed, Scopus, and Web of Science databases were queried on March 2024 for studies evaluating adjuvant therapies in patients treated with surgery for localized ACC (PROSPERO: CRD42024512849). The endpoints of interest were overall survival (OS) and recurrence-free survival (RFS). Hazard ratios (HR) with 95% confidence intervals (95%CI) were pooled in a random-effects model meta-analysis. One randomized controlled trial (n = 91) and eleven retrospective studies (n = 4,515) were included. Adjuvant mitotane therapy was associated with improved RFS (HR: 0.63, 95%CI: 0.44-0.92, p = 0.016), while adjuvant RT did not reach conventional levels of statistical significance (HR:0.79, 95%CI:0.58-1.06, p = 0.11). Conversely, Adjuvant RT was associated with improved OS (HR:0.69, 95%CI:0.58-0.83, p<0.001), whereas adjuvant mitotane did not (HR: 0.76, 95%CI: 0.57-1.02, p = 0.07). In the subgroup analyses, adjuvant mitotane was associated with better OS (HR:0.46, 95%CI: 0.30-0.69, p < 0.001) and RFS (HR:0.56, 95%CI: 0.32-0.98, p = 0.04) in patients with negative surgical margin. Both adjuvant RT and mitotane were found to be associated with improved oncologic outcomes in patients treated with adrenalectomy for localised ACC. While adjuvant RT significantly improved OS in general population, mitotane appears as an especially promising treatment option in patients with negative surgical margin. These data can support the shared decision-making process, better understanding of the risks, benefits, and effectiveness of these therapies is still needed to guide tailored management of each individual patient.
Collapse
Affiliation(s)
- Ichiro Tsuboi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Shimane University Faculty of Medicine, Shimane, Japan; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mehdi Kardoust Parizi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Akihiro Matsukawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Stefano Mancon
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Marcin Miszczyk
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Collegium Medicum - Faculty of Medicine, WSB University, Dąbrowa Górnicza, Poland
| | - Robert J Schulz
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tamás Fazekas
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Semmelweis University, Budapest, Hungary
| | - Anna Cadenar
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Unit of Oncologic Minimally Invasive Urology and Andrology, Department of Experimental and Clinical Medicine, Careggi Hospital, University of Florence, Florence, Italy
| | - Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Tatsushi Kawada
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Katayama
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takehiro Iwata
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kensuke Bekku
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koichiro Wada
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Mesut Remzi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, Quebec, Canada
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia; Department of Urology, University of Texas Southwestern, Dallas, TX; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic; Division of Urology, Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria; Research center of Evidence Medicine, Urology department Tabriz University of Medical Sciences, Tabriz, Iran; Department of Urology, Semmelweis University, Budapest, Hungary; Department of Urology, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
4
|
Arakawa Y, Elloumi F, Varma S, Khandagale P, Jo U, Kumar S, Roper N, Reinhold WC, Robey RW, Takebe N, Gottesman MM, Thomas CJ, Boeva V, Berruti A, Abate A, Tamburello M, Sigala S, Hantel C, Weigand I, Wierman ME, Kiseljak-Vassiliades K, Del Rivero J, Pommier Y. A Database Tool Integrating Genomic and Pharmacologic Data from Adrenocortical Carcinoma Cell Lines, PDX, and Patient Samples. CANCER RESEARCH COMMUNICATIONS 2024; 4:2384-2398. [PMID: 39162009 PMCID: PMC11389377 DOI: 10.1158/2767-9764.crc-24-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/07/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare and highly heterogeneous disease with a notably poor prognosis due to significant challenges in diagnosis and treatment. Emphasizing on the importance of precision medicine, there is an increasing need for comprehensive genomic resources alongside well-developed experimental models to devise personalized therapeutic strategies. We present ACC_CellMinerCDB, a substantive genomic and drug sensitivity database (available at https://discover.nci.nih.gov/acc_cellminercdb) comprising ACC cell lines, patient-derived xenografts, surgical samples, and responses to more than 2,400 drugs examined by the NCI and National Center for Advancing Translational Sciences. This database exposes shared genomic pathways among ACC cell lines and surgical samples, thus authenticating the cell lines as research models. It also allows exploration of pertinent treatment markers such as MDR-1, SOAT1, MGMT, MMR, and SLFN11 and introduces the potential to repurpose agents like temozolomide for ACC therapy. ACC_CellMinerCDB provides the foundation for exploring larger preclinical ACC models. SIGNIFICANCE ACC_CellMinerCDB, a comprehensive database of cell lines, patient-derived xenografts, surgical samples, and drug responses, reveals shared genomic pathways and treatment-relevant markers in ACC. This resource offers insights into potential therapeutic targets and the opportunity to repurpose existing drugs for ACC therapy.
Collapse
Affiliation(s)
- Yasuhiro Arakawa
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fathi Elloumi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sudhir Varma
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Prashant Khandagale
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ukhyun Jo
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Suresh Kumar
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nitin Roper
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William C. Reinhold
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Naoko Takebe
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Valentina Boeva
- Department of Computer Science, Institute for Machine Learning, ETH Zurich, Zurich, Switzerland
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Medical Oncology Unit, University of Brescia, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, and University of Zurich, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Isabel Weigand
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Margaret E. Wierman
- Department of Medicine-Endocrinology/Metabolism/Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Katja Kiseljak-Vassiliades
- Department of Medicine-Endocrinology/Metabolism/Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
5
|
Wu K, Liu Z, Liang J, Zhu Y, Wang X, Li X. Discovery of a glucocorticoid receptor (GR) activity signature correlates with immune cell infiltration in adrenocortical carcinoma. J Immunother Cancer 2023; 11:e007528. [PMID: 37793855 PMCID: PMC10551943 DOI: 10.1136/jitc-2023-007528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is a rare and highly aggressive endocrine malignancy, of which >40% present with glucocorticoid excess. Glucocorticoids and glucocorticoid receptor (GR) signaling have long been thought to suppress immunity and promote tumor progression by acting on immune cells. Here, we provide new insights into the interaction between GR signaling activity and the immune signature of ACC as a potential explanation for immune escape and resistance to immunotherapy. METHODS First, GR immunohistochemical staining and immunofluorescence analysis of tumor-infiltrating lymphocyte (CD4 T, CD8 T cells, natural killer (NK) cells, dendritic cells and macrophages) were performed in 78 primary ACC tissue specimens. Quantitative data of immune cell infiltration in ACC were correlated with clinical characteristics. Second, we discovered a GR activity signature (GRsig) using GR-targeted gene networks derived from global gene expression data of primary ACC. Finally, we identified two GRsig-related subtypes based on the GRsig and assessed the differences in immune characteristics and prognostic stratification between the two subtypes. RESULTS GR was expressed in 90% of the ACC tumors, and CD8+ cytotoxic T lymphocytes were the most common infiltrating cell type in ACC specimens (88%, 8.6 cells/high power field). GR expression positively correlated with CD8+ T cell (Phi=0.342, p<0.001), CD4+ T cell (Phi=0.280, p<0.001), NK cell (Phi=0.280, p<0.001), macrophage (Phi=0.285, p<0.001), and dendritic cell (Phi=0.397, p<0.001) infiltration. Clustering heatmap analysis also displayed high immune cell infiltration in GR high-expressing tumors and low immune cell infiltration in GR-low tumors. High GR expression and high immune cell infiltration were significantly associated with better survival. Glucocorticoid excess is associated with low immune cell abundance and unfavorable prognosis. A GRsig comprizing n=34 GR-associated genes was derived from Gene Expression Omnibus/The Cancer Genome Atlas (TCGA) data sets and used to define two GRsig-related subtypes in the TCGA cohort. We demonstrated distinct differences in the immune landscape and clinical outcomes between the two subtypes. CONCLUSION GR expression positively correlates with tumor-infiltrating immune cells in ACC. The GRsig could serve as a prognostic biomarker and may be helpful for prognosis prediction and response to immunotherapy. Consequently, targeting the GR signaling pathway might be pivotal and should be investigated in clinical studies.
Collapse
Affiliation(s)
- Kan Wu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhihong Liu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayu Liang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuchun Zhu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xianding Wang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiang Li
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Dedhia PH, Sivakumar H, Rodriguez MA, Nairon KG, Zent JM, Zheng X, Jones K, Popova LV, Leight JL, Skardal A. A 3D adrenocortical carcinoma tumor platform for preclinical modeling of drug response and matrix metalloproteinase activity. Sci Rep 2023; 13:15508. [PMID: 37726363 PMCID: PMC10509170 DOI: 10.1038/s41598-023-42659-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
Adrenocortical carcinoma (ACC) has a poor prognosis, and no new drugs have been identified in decades. The absence of drug development can partly be attributed to a lack of preclinical models. Both animal models and 2D cell cultures of ACC fail to accurately mimic the disease, as animal physiology is inherently different than humans, and 2D cultures fail to represent the crucial 3D architecture. Organoids and other small 3D in vitro models of tissues or tumors can model certain complexities of human in vivo biology; however, this technology has largely yet to be applied to ACC. In this study, we describe the generation of 3D tumor constructs from an established ACC cell line, NCI-H295R. NCI-H295R cells were encapsulated to generate 3D ACC constructs. Tumor constructs were assessed for biomarker expression, viability, proliferation, and cortisol production. In addition, matrix metalloproteinase (MMP) functionality was assessed directly using fluorogenic MMP-sensitive biosensors and through infusion of NCI-H295R cells into a metastasis-on-a-chip microfluidic device platform. ACC tumor constructs showed expression of biomarkers associated with ACC, including SF-1, Melan A, and inhibin α. Treatment of ACC tumor constructs with chemotherapeutics demonstrated decreased drug sensitivity compared to 2D cell culture. Since most tumor cells migrate through tissue using MMPs to break down extracellular matrix, we validated the utility of ACC tumor constructs by integrating fluorogenic MMP-sensitive peptide biosensors within the tumor constructs. Lastly, in our metastasis-on-a-chip device, NCI-H295R cells successfully engrafted in a downstream lung cell line-based construct, but invasion distance into the lung construct was decreased by MMP inhibition. These studies, which would not be possible using 2D cell cultures, demonstrated that NCI-H295R cells secreted active MMPs that are used for invasion in 3D. This work represents the first evidence of a 3D tumor constructs platform for ACC that can be deployed for future mechanistic studies as well as development of new targets for intervention and therapies.
Collapse
Affiliation(s)
- Priya H Dedhia
- Division of Surgical Oncology, The Ohio State University and Arthur G. James Comprehensive Cancer Center, 816 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- Translational Therapeutics Program, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA.
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, USA.
| | - Hemamylammal Sivakumar
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Marco A Rodriguez
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Kylie G Nairon
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Joshua M Zent
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Xuguang Zheng
- Division of Surgical Oncology, The Ohio State University and Arthur G. James Comprehensive Cancer Center, 816 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Katie Jones
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Liudmila V Popova
- Division of Surgical Oncology, The Ohio State University and Arthur G. James Comprehensive Cancer Center, 816 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Jennifer L Leight
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, USA.
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- Cancer Biology Program, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA.
| | - Aleksander Skardal
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, USA.
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, 886 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- Cancer Biology Program, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
7
|
Dedhia P, Sivakumar H, Rodriguez MA, Nairon KG, Zent JM, Zheng X, Jones K, Popova L, Leight JL, Skardal A. A 3D adrenocortical carcinoma tumor platform for preclinical modeling of drug response and matrix metalloproteinase activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525287. [PMID: 36747748 PMCID: PMC9900758 DOI: 10.1101/2023.01.24.525287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Adrenocortical carcinoma (ACC) has a poor prognosis, and no new drugs have been identified in decades. The absence of drug development can partly be attributed to a lack of preclinical models. Both animal models and 2D cell cultures of ACC fail to accurately mimic the disease, as animal physiology is inherently different than humans, and 2D cultures fail to represent the crucial 3D architecture. Organoids and other small 3D in vitro models of tissues or tumors can model certain complexities of human in vivo biology; however, this technology has largely yet to be applied to ACC. In this study, we describe the generation of 3D tumor constructs from an established ACC cell line, NCI-H295R. NCI-H295R cells were encapsulated to generate 3D ACC constructs. Tumor constructs were assessed for biomarker expression, viability, proliferation, and cortisol production. In addition, matrix metalloproteinase (MMP) functionality was assessed directly using fluorogenic MMP-sensitive biosensors and through infusion of NCI-H295R cells into a metastasis-on-a-chip microfluidic device platform. ACC tumor constructs showed expression of biomarkers associated with ACC, including SF-1, Melan A, and inhibin alpha. Treatment of ACC tumor constructs with chemotherapeutics demonstrated decreased drug sensitivity compared to 2D cell culture. Since most tumor cells migrate through tissue using MMPs to break down extracellular matrix, we validated the utility of ACC tumor constructs by integrating fluorogenic MMP-sensitive peptide biosensors within the tumor constructs. Lastly, in our metastasis-on-a-chip device, NCI-H295R cells successfully engrafted in a downstream lung cell line-based construct, but invasion distance into the lung construct was decreased by MMP inhibition. These studies, which would not be possible using 2D cell cultures, demonstrated that NCI-H295R cells secreted active MMPs that are used for invasion in 3D. This work represents the first evidence of a 3D tumor constructs platform for ACC that can be deployed for future mechanistic studies as well as development of new targets for intervention and therapies.
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Adrenocortical carcinoma (ACC) is a rare, aggressive disease with a paucity of data and great variability between published studies regarding its treatment. This review provides information on current clinical management and oncological and endocrine outcomes. RECENT FINDINGS Complete surgical resection is the only potentially curative treatment for adrenocortical carcinoma (ACC). Adjuvant mitotane treatment is recommended in patients with favourable/intermediate prognosis. As part of the endocrine follow-up, steroid hormones and thyroid hormones may be decreased or increased and may need to be substituted or suppressed. Recurrences are common. If the disease-free interval is more than 12 months, surgery is a treatment if complete resection is feasible. In advanced/metastatic ACC patients, the prognosis is poor. Mitotane monotherapy is only appropriate for patients with low tumour burden and indolent disease. Patients with unfavourable prognosis should be treated with aggressive cytotoxic therapy. Patients requiring third-line treatment should be considered for clinical trials. Immunotherapy and targeted therapy are currently being investigated, but have so far yielded only unsatisfactory results. SUMMARY There is scarce evidence for the treatment of ACC, which often complicates clinical decision-making. Patients who progress on EDP-M should be treated in clinical trials.
Collapse
|
9
|
Sigala S, Rossini E, Abate A, Tamburello M, Bornstein SR, Hantel C. An update on adrenocortical cell lines of human origin. Endocrine 2022; 77:432-437. [PMID: 35764904 PMCID: PMC9385758 DOI: 10.1007/s12020-022-03112-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare, heterogenous and highly malignant disease. Management of ACC is dependent on disease stage with complete surgical resection as the only potentially curative option. However, advanced, un-resectable, metastatic stages and also recurrences often require systemic treatments, which are unfortunately nowadays still unsatisfactory. The scarcity of preclinical models reflecting patient heterogeneities and furthermore drug-resistant phenotypes, has hampered the progress and development of new therapies in recent years. In this review, we provide an overview on the classical models and substantial progress which has been made over the last years in context of this aggressive disease.
Collapse
Affiliation(s)
- Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Stefan R Bornstein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, London, WC2R 2LS, UK
- Center for Regenerative Therapies, Technische Universität Dresden, 01307, Dresden, Germany
- Paul-Langerhans-Institute Dresden, Helmholtz Center Munich, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, 01307, Dresden, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland.
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany.
| |
Collapse
|
10
|
Alyateem G, Nilubol N. Current Status and Future Targeted Therapy in Adrenocortical Cancer. Front Endocrinol (Lausanne) 2021; 12:613248. [PMID: 33732213 PMCID: PMC7957049 DOI: 10.3389/fendo.2021.613248] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis. The current treatment standards include complete surgical resection for localized resectable disease and systemic therapy with mitotane alone or in combination with etoposide, doxorubicin, and cisplatin in patients with advanced ACC. However, the efficacy of systemic therapy in ACC is very limited, with high rates of toxicities. The understanding of altered molecular pathways is critically important to identify effective treatment options that currently do not exist. In this review, we discuss the results of recent advanced in molecular profiling of ACC with the focus on dysregulated pathways from various genomic and epigenetic dysregulation. We discuss the potential translational therapeutic implication of molecular alterations. In addition, we review and summarize the results of recent clinical trials and ongoing trials.
Collapse
|
11
|
Zhu J, Zheng Z, Shen J, Lian X, Miao Z, Shen J, Zhang F. Efficacy of adjuvant radiotherapy for treatment of adrenocortical carcinoma: a retrospective study and an updated meta-analysis. Radiat Oncol 2020; 15:118. [PMID: 32448148 PMCID: PMC7245885 DOI: 10.1186/s13014-020-01533-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Background Adrenocortical carcinoma (ACC) is a rare and highly aggressive malignancy. Surgical resection is the standard treatment for localized ACC, but the local recurrence remains high. Adjuvant radiation (ART) has been proposed as a means to reduce recurrence rates in ACC after surgery with conflicting results from nonrandomized studies. We performed a retrospective study and a meta-analysis to determine the impact of ART on survival outcomes. Methods A retrospective study of the adrenocortical cancer database in Peking Union Medical College was conducted. We selected postoperative ACC patients with or without ART. A meta-analysis is also performed to compare the outcomes between ART and only surgical resection in ACC patients. The obtained data were analyzed using SPSS 23.0 and Stata 15.0 statistical software. Differences between two groups were compared using the log-rank test for retrospective analysis and estimated by calculating the odds ratio (OR) and 95% confidence interval (CI) for meta-analysis. Results Of a total of 75 patients available in the database, 12 patients underwent postoperative ART and were matched one to one to patients with only surgical resection. There was no significant difference on overall survival between ART group and control group (log-rank P = 0.149). Locoregional recurrence was diagnosed in 2 of the ART group, and in 4 of the control group (P = 0.64). A total of 238 participants were selected for the meta-analysis, of which 111 and 127 patients underwent ART after surgical resection and only surgical resection, respectively. Overall survival is significantly higher in ART group, with an odds ratio (OR) of 2.41 (95% CI of 1.33, 4.38; P = 0.004). Besides, meta-analysis significantly favored ART for locoregional recurrence-free survival and disease-free survival, with an OR of 4.08 and 2.27, respectively. Conclusions Our results show that compared to only surgical resection, ART is an effective postoperative treatment for ACC.
Collapse
Affiliation(s)
- Jiawei Zhu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Ziye Zheng
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jing Shen
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xin Lian
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Zheng Miao
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jie Shen
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Fuquan Zhang
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academe of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
12
|
Rizk-Rabin M, Chaoui-Ibadioune S, Vaczlavik A, Ribes C, Polak M, Ragazzon B, Bertherat J. Link between steroidogenesis, the cell cycle, and PKA in adrenocortical tumor cells. Mol Cell Endocrinol 2020; 500:110636. [PMID: 31678420 DOI: 10.1016/j.mce.2019.110636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 10/09/2019] [Accepted: 10/26/2019] [Indexed: 02/04/2023]
Abstract
Adrenocortical tumors (ACTs) frequently cause steroid excess and present cell-cycle dysregulation. cAMP/PKA signaling is involved in steroid synthesis and play a role in cell-cycle regulation. We investigated, by cell synchronization in the different phases of the cell-cycle, the control of steroidogenesis and the contribution of PKA in adrenocortical cells (H295R and culture of primary pigmented nodular adrenocortical disease cells). Cells showed increased steroidogenesis and a maximal PKA activity at G2 phase, and a reduction at G1 phase. PRKACA overexpression, or cAMP stimulation, enhanced PKA activity and induced steroidogenesis in all synchronized groups but is not sufficient to drive cell-cycle progression. PRKAR1A inactivation enhanced PKA activity and induced STAR gene expression, only in cells in G1, and triggered cell-cycle progression in all groups. These findings provide evidence for a tight association between steroidogenesis and cell-cycle in ACTs. Moreover, PRKAR1A is essential for mediating the function of PKA activity on both steroidogenesis and cell-cycle progression in adrenocortical cells.
Collapse
Affiliation(s)
- Marthe Rizk-Rabin
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France.
| | | | - Anna Vaczlavik
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France
| | - Christopher Ribes
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France
| | - Michel Polak
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France; Hopital Necker Enfants Maladies, Department of Endocrinology, Paris, France
| | - Bruno Ragazzon
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France
| | - Jerôme Bertherat
- Institut Cochin, U1016, CNRS (UMR 8104), Université Paris Descartes, Paris, France; Hôpital Cochin, Department of Endocrinology. Center for Rare Adrenal Diseases, Paris, France
| |
Collapse
|
13
|
Cerquetti L, Bucci B, Carpinelli G, Lardo P, Proietti A, Saporito R, Rindi G, Petrangeli E, Toscano V, Stigliano A. Antineoplastic Effect of a Combined Mitotane Treatment/Ionizing Radiation in Adrenocortical Carcinoma: A Preclinical Study. Cancers (Basel) 2019; 11:cancers11111768. [PMID: 31717612 PMCID: PMC6895792 DOI: 10.3390/cancers11111768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/24/2019] [Accepted: 11/07/2019] [Indexed: 11/17/2022] Open
Abstract
Mitotane (MTT) is an adrenolytic drug used in adjuvant and advanced treatments of adrenocortical carcinoma (ACC). Ionizing radiation (IR) is also used in adrenal cancer treatment, even though its biological action remains unknown. To provide a reliable in vivo preclinical model of ACC, we used mouse xenografts bearing human ACC to test the effects of MTT and IR alone and in combination. We evaluated tumor growth inhibition by the RECIST criteria and analyzed the cell cycle by flow cytometry (FCM). In the xenograft ACC model treated with MTT/IR in combination, we observed a marked inhibition of tumor growth, with strong tumor regression (p < 0.0001) compared to MTT and IR given alone (p < 0.05). The MTT results confirm its antisteroidogenic activity (p < 0.05) in the xenograft ACC model, revealing its ability to render cancer cells more prone to radiotherapy treatment. In addition, to explain the biological effect of these treatments on the Mismatch Repair System (MMR), we interfered with the MSH2 gene expression in untreated and MTT/IR-treated H295R and SW13 cell lines. Moreover, we observed that upon treatment with MTT/IR to induce DNA damage, MSH2 gene inhibition in both the H295R and SW13 cell lines did not allow DNA damage repair, thus inducing cell death. In conclusion, MTT seems to have a radiosensitizing property and, when given in combination with IR, is able to promote neoplastic growth inhibition, leading to a significant reduction in tumor size due to cell death.
Collapse
Affiliation(s)
- Lidia Cerquetti
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (L.C.); (P.L.); (V.T.)
| | - Barbara Bucci
- UOC Pathological Clinic San Pietro Hospital Fatebenefratelli, 00189 Rome, Italy; (B.B.); (R.S.)
| | - Giulia Carpinelli
- Department of Cellular Biology and Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Pina Lardo
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (L.C.); (P.L.); (V.T.)
| | - Antonella Proietti
- Diagnostic of Laboratory Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| | - Raffaele Saporito
- UOC Pathological Clinic San Pietro Hospital Fatebenefratelli, 00189 Rome, Italy; (B.B.); (R.S.)
| | - Guido Rindi
- Pathology Unit, University Catholic, 00168 Rome, Italy;
| | - Elisa Petrangeli
- CNR, Institute of Molecular Biology and Pathology, 00185 Rome, Italy;
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Vincenzo Toscano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (L.C.); (P.L.); (V.T.)
| | - Antonio Stigliano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (L.C.); (P.L.); (V.T.)
- Correspondence: ; Tel.: +39-6-3377-4784; Fax: +39-6-3377-6327
| |
Collapse
|
14
|
Abstract
Adrenocortical carcinoma (ACC) is an uncommon and heterogeneous disease and may present differently in children and adults. Management of ACC is dependent on disease stage and complete surgical resection is the only potentially curative treatment. The first and most extensively used adrenocortical cancer cell line, as model system to examine mechanisms controlling normal and pathologic function of adrenal cortex, was initially isolated in 1980. Although NCI-H295 maintained steroid capabilities and adrenocortical characteristics, the lack of new cell lines and animal models of ACC has hampered the progress and development of new therapies. In this review we provide description of cellular and patient-derived tumor xenograft (PDTX) models of ACC generated for the elucidation of the underlying pathogenic mechanisms and preclinical functional studies for this aggressive disease.
Collapse
|
15
|
Abstract
Adrenocortical carcinoma (ACC) is a rare, aggressive, and frequently deadly cancer. Up to 75% of all patients will eventually develop metastatic disease, and our current medical therapies for ACC provide limited - if any - survival benefit. These statistics highlight a crucial need for novel approaches. Recent studies performing comprehensive molecular profiling on ACC have illuminated that ACC is comprised of three clinically distinct molecular subtypes, bearing differential regulation of cell cycle, epigenetics, Wnt/β-catenin signaling, PKA signaling, steroidogenesis and immune cell biology. Furthermore, these studies have spurred the development of molecular subtype-based biomarkers, contextualized outcomes of recent clinical trials, and advanced our understanding of the underlying biology of adrenocortical homeostasis and cancer. In this review, we describe these findings and their implications for new strategies to apply targeted therapies to ACC.
Collapse
|
16
|
Nicolson NG, Korah R, Carling T. Adrenocortical cancer cell line mutational profile reveals aggressive genetic background. J Mol Endocrinol 2019; 62:179-186. [PMID: 30870809 DOI: 10.1530/jme-18-0262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/19/2022]
Abstract
Adrenocortical carcinomas are rare tumors with poor prognosis and limited treatment options. Although widely used as in vitro models to test novel therapeutic strategies, the adrenocortical carcinoma-derived cell lines NCI-H295R and SW-13 have only partially been described genetically. Our aim was to characterize the mutational landscape of these cells to improve their experimental utility and map them to clinical subtypes of adrenocortical carcinoma. Genomic DNA from NCI-H295R and SW-13 cells was subjected to whole-exome sequencing. Variants were filtered for non-synonymous mutations and curated for validated adrenocortical and pan-cancer driver gene mutations. Genes mutated in the cell lines were mapped using gene ontology and protein pathway tools to determine signaling effects and compared to mutational and clinical characteristics of 92 adrenocortical carcinoma cases from The Cancer Genome Atlas. NCI-H295R and SW-13 cells carried 1325 and 1836 non-synonymous variants, respectively. Of these, 61 and 76 were known cancer driver genes, of which 32 were shared between cell lines. Variant interaction analyses demonstrated dominant TP53 dysregulation in both cell lines complemented by distinct WNT (NCI-H295R) and chromatin remodeling (SW-13) pathway perturbations. Both cell lines genetically resemble more aggressive adrenocortical carcinomas with worse prognosis, for which development of targeted therapies is most critical. Careful incorporation of the genetic landscapes outlined in this study will further the in vitro utility of these cell lines in testing for novel therapeutic approaches for adrenocortical malignancy.
Collapse
Affiliation(s)
- Norman G Nicolson
- Department of Surgery, Yale School of Medicine, Yale Endocrine Neoplasia Laboratory, New Haven, Connecticut, USA
| | - Reju Korah
- Department of Surgery, Yale School of Medicine, Yale Endocrine Neoplasia Laboratory, New Haven, Connecticut, USA
| | - Tobias Carling
- Department of Surgery, Yale School of Medicine, Yale Endocrine Neoplasia Laboratory, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Fassnacht M, Dekkers O, Else T, Baudin E, Berruti A, de Krijger R, Haak H, Mihai R, Assie G, Terzolo M. European Society of Endocrinology Clinical Practice Guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol 2018; 179:G1-G46. [PMID: 30299884 DOI: 10.1530/eje-18-0608] [Citation(s) in RCA: 546] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare and in most cases steroid hormone-producing tumor with variable prognosis. The purpose of these guidelines is to provide clinicians with best possible evidence-based recommendations for clinical management of patients with ACC based on the GRADE (Grading of Recommendations Assessment, Development and Evaluation) system. We predefined four main clinical questions, which we judged as particularly important for the management of ACC patients and performed systematic literature searches: (A) What is needed to diagnose an ACC by histopathology? (B) Which are the best prognostic markers in ACC? (C) Is adjuvant therapy able to prevent recurrent disease or reduce mortality after radical resection? (D) What is the best treatment option for macroscopically incompletely resected, recurrent or metastatic disease? Other relevant questions were discussed within the group. Selected Recommendations: (i) We recommend that all patients with suspected and proven ACC are discussed in a multidisciplinary expert team meeting. (ii) We recommend that every patient with (suspected) ACC should undergo careful clinical assessment, detailed endocrine work-up to identify autonomous hormone excess and adrenal-focused imaging. (iii) We recommend that adrenal surgery for (suspected) ACC should be performed only by surgeons experienced in adrenal and oncological surgery aiming at a complete en bloc resection (including resection of oligo-metastatic disease). (iv) We suggest that all suspected ACC should be reviewed by an expert adrenal pathologist using the Weiss score and providing Ki67 index. (v) We suggest adjuvant mitotane treatment in patients after radical surgery that have a perceived high risk of recurrence (ENSAT stage III, or R1 resection, or Ki67 >10%). (vi) For advanced ACC not amenable to complete surgical resection, local therapeutic measures (e.g. radiation therapy, radiofrequency ablation, chemoembolization) are of particular value. However, we suggest against the routine use of adrenal surgery in case of widespread metastatic disease. In these patients, we recommend either mitotane monotherapy or mitotane, etoposide, doxorubicin and cisplatin depending on prognostic parameters. In selected patients with a good response, surgery may be subsequently considered. (vii) In patients with recurrent disease and a disease-free interval of at least 12 months, in whom a complete resection/ablation seems feasible, we recommend surgery or alternatively other local therapies. Furthermore, we offer detailed recommendations about the management of mitotane treatment and other supportive therapies. Finally, we suggest directions for future research.
Collapse
Affiliation(s)
- Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Olaf Dekkers
- Department of Clinical Epidemiology
- Department of Clinical Endocrinology and Metabolism, Leiden University Medical Centre, Leiden, the Netherlands
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Tobias Else
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric Baudin
- Endocrine Oncology and Nuclear Medicine, Institut Gustave Roussy, Villejuif, France
- INSERM UMR 1185, Faculté de Médecine, Le Kremlin-Bicêtre, Université Paris Sud, Paris, France
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Medical Oncology, University of Brescia at ASST Spedali Civili, Brescia, Italy
| | - Ronald de Krijger
- Department of Pathology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Pathology, Reinier de Graaf Hospital, Delft, the Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Harm Haak
- Department of Internal Medicine, Máxima Medical Centre, Eindhoven/Veldhoven, the Netherlands
- Maastricht University, CAPHRI School for Public Health and Primary Care, Ageing and Long-Term Care, Maastricht, the Netherlands
- Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Radu Mihai
- Department of Endocrine Surgery, Churchill Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Guillaume Assie
- Department of Endocrinology, Reference Center for Rare Adrenal Diseases, Reference Center dor Rare Adrenal Cancers, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Massimo Terzolo
- Department of Clinical and Biological Sciences, Internal Medicine, San Luigi Hospital, University of Turin, Orbassano, Italy
| |
Collapse
|
18
|
Tamamori-Adachi M, Koga A, Susa T, Fujii H, Tsuchiya M, Okinaga H, Hisaki H, Iizuka M, Kitajima S, Okazaki T. DNA damage response induced by Etoposide promotes steroidogenesis via GADD45A in cultured adrenal cells. Sci Rep 2018; 8:9636. [PMID: 29941883 PMCID: PMC6018231 DOI: 10.1038/s41598-018-27938-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 06/12/2018] [Indexed: 11/17/2022] Open
Abstract
Glucocorticoid production is regulated by adrenocorticotropic hormone (ACTH) via the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway in the adrenal cortex, but the changes in steroidogenesis associated with aging are unknown. In this study, we show that cell-autonomous steroidogenesis is induced by non-ACTH- mediated genotoxic stress in human adrenocortical H295R cells. Low-dose etoposide (EP) was used to induce DNA damage as a genotoxic stress, leading to cellular senescence. We found that steroidogenesis was promoted in cells stained with γH2AX, a marker of DNA damaged cells. Among stress-associated and p53-inducible genes, the expression of GADD45A and steroidogenesis-related genes was significantly upregulated. Immunofluorescence analysis revealed that GADD45A accumulated in the nuclei. Metabolite assay using cultured media showed that EP-treated cells were induced to produce and secrete considerable amounts of glucocorticoid. Knockdown of GADD45A using small interfering RNA markedly inhibited the EP-induced upregulation of steroidogenesis-related gene expression, and glucocorticoid production. A p38MAPK inhibitor, but not a PKA inhibitor, suppressed EP-stimulated steroidogenesis. These results suggest that DNA damage itself promotes steroidogenesis via one or more unprecedented non-ACTH-mediated pathway. Specifically, GADD45A plays a crucial role in the steroidogenic processes triggered by EP-stimulated genotoxic stress. Our study sheds new light on an alternate mechanism of steroidogenesis in the adrenal cortex.
Collapse
Affiliation(s)
- Mimi Tamamori-Adachi
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Akane Koga
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.,Department of Practical Pharmacy, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Takao Susa
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Hiroko Fujii
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.,Department of General Medicine, National Defense Medical College, 3-2, Namiki, Tokorozawa City, Saitama, 359-8513, Japan
| | - Masao Tsuchiya
- Department of Practical Pharmacy, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Hiroko Okinaga
- Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Harumi Hisaki
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Masayoshi Iizuka
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Shigetaka Kitajima
- Department of Biochemical Genetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8605, Japan
| | - Tomoki Okazaki
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| |
Collapse
|
19
|
Srougi V, de Bessa J, Tanno FY, Ferreira AM, Hoff AO, Bezerra JE, Almeida CM, Almeida MQ, Mendonça BB, Nahas WC, Chambô JL, Srougi M, Fragoso MCBV. Adjuvant radiotherapy for the primary treatment of adrenocortical carcinoma: Are we offering the best? Int Braz J Urol 2017; 43:841-848. [PMID: 28727379 PMCID: PMC5678514 DOI: 10.1590/s1677-5538.ibju.2017.0095] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/09/2017] [Indexed: 01/13/2023] Open
Abstract
Purpose: To evaluate the role of ARDT after surgical resection of ACC. Materials and Methods: Records of patients from our institutional ACC database were retrospectively assessed. A paired comparison analysis was used to evaluate the oncological outcomes between patients treated with surgery followed by ARDT or surgery only (control). The endpoints were LRFS, RFS, and OS. A systematic review of the literature and meta-analysis was also performed to evaluate local recurrence of ACC when ARDT was used. Results: Ten patients were included in each Group. The median follow-up times were 32 months and 35 months for the ARDT and control Groups, respectively. The results for LRFS (p=0.11), RFS (p=0.92), and OS (p=0.47) were similar among subsets. The mean time to present with local recurrence was significantly longer in the ARDT group compared with the control Group (419±206 days vs. 181±86 days, respectively; p=0.03). ARDT was well tolerated by the patients; there were no reports of late toxicity. The meta-analysis, which included four retrospective series, revealed that ARDT had a protective effect on LRFS (HR=0.4; CI=0.17-0.94). Conclusions: ARDT may reduce the chance and prolong the time to ACC local recurrence. However, there were no benefits for disease recurrence control or overall survival for patients who underwent this complementary therapy.
Collapse
Affiliation(s)
- Victor Srougi
- Divisão de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Jose de Bessa
- Divisão de Urologia, Universidade de Feira de Santana, BA, Brasil
| | - Fabio Y Tanno
- Divisão de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Amanda M Ferreira
- Unidade de Suprerrenal da Divisão de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, Brasil
| | - Ana O Hoff
- Divisão de Endocrinologia do Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - João E Bezerra
- Divisão de Endocrinologia do Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Cristiane M Almeida
- Divisão de Radioterapia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Madson Q Almeida
- Unidade de Suprerrenal da Divisão de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, Brasil.,Divisão de Endocrinologia do Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Berenice B Mendonça
- Unidade de Suprerrenal da Divisão de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, Brasil
| | - William C Nahas
- Divisão de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Jose L Chambô
- Divisão de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Miguel Srougi
- Divisão de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Maria C B V Fragoso
- Unidade de Suprerrenal da Divisão de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, Brasil.,Divisão de Endocrinologia do Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| |
Collapse
|
20
|
Stigliano A, Cerquetti L, Lardo P, Petrangeli E, Toscano V. New insights and future perspectives in the therapeutic strategy of adrenocortical carcinoma (Review). Oncol Rep 2017; 37:1301-1311. [PMID: 28184938 DOI: 10.3892/or.2017.5427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/20/2016] [Indexed: 11/06/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with an incidence ranging from 0.7 to 2.0 cases/million people per year. Hypercortisolism represents the most common clinical presentation in many patients although, less frequently, some ACC secreting androgens and estrogens are even more pathognomonic compared to cortisol secretion. Currently, radical surgery, when feasible, is still the only curative therapy. Mitotane, an adrenolytic drug, is used in the adjuvant setting and in combination with chemotherapy drugs in metastatic disease. The use of radiotherapy remains controversial, being indicated only in selected cases. New targeted therapies, such as insulin growth factor-1 (IGF-1), mammalian-target of rapamycin (m-TOR), vascular endothelial growth factor (VEGF) inhibitors and others, have recently been investigated with disappointing clinical results. The partial effectiveness of current treatments mandates the need for new therapeutic strategies against this tumor.
Collapse
Affiliation(s)
- Antonio Stigliano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Lidia Cerquetti
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Pina Lardo
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Elisa Petrangeli
- CNR, Institute of Molecular Biology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Vincenzo Toscano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
21
|
Hirsutismo y amenorrea en carcinoma adrenocortical oncocítico hormonalmente activo. Aportación de un caso y revisión de la literatura. Rev Int Androl 2017. [DOI: 10.1016/j.androl.2016.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Srdic-Rajic T, Tisma-Miletic N, Cavic M, Kanjer K, Savikin K, Galun D, Konic-Ristic A, Zoranovic T. Sensitization of K562 Leukemia Cells to Doxorubicin by theViscum albumExtract. Phytother Res 2015; 30:485-95. [DOI: 10.1002/ptr.5554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/02/2015] [Accepted: 11/25/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Tatjana Srdic-Rajic
- Department of Experimental Pharmacology; Institute for Oncology and Radiology of Serbia; Belgrade Serbia
| | - Nevena Tisma-Miletic
- Department of Experimental Pharmacology; Institute for Oncology and Radiology of Serbia; Belgrade Serbia
| | - Milena Cavic
- Department of Experimental Pharmacology; Institute for Oncology and Radiology of Serbia; Belgrade Serbia
| | - Ksenija Kanjer
- Department of Experimental Pharmacology; Institute for Oncology and Radiology of Serbia; Belgrade Serbia
| | - Katarina Savikin
- Institute for Medicinal Plant Research ‘Dr Josif Pančić’; Belgrade Serbia
| | - Danijel Galun
- University Clinic for Digestive Surgery; Clinical center Serbia; Belgrade Serbia
- Belgrade University Medical School; Belgrade Serbia
| | - Aleksandra Konic-Ristic
- Institute for Medical Research, Center of Research Excellence in Nutrition and Metabolism; Belgrade University; Belgrade Serbia
| | - Tamara Zoranovic
- Department of Experimental Pharmacology; Institute for Oncology and Radiology of Serbia; Belgrade Serbia
- Max Plank Institute for Infection Biology; Berlin Germany
| |
Collapse
|
23
|
Adjuvant radiation therapy improves local control after surgical resection in patients with localized adrenocortical carcinoma. Int J Radiat Oncol Biol Phys 2015; 92:252-9. [PMID: 25754631 DOI: 10.1016/j.ijrobp.2015.01.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/08/2014] [Accepted: 01/09/2015] [Indexed: 02/03/2023]
Abstract
PURPOSE Adrenocortical carcinoma (ACC) is a rare malignancy known for high rates of local recurrence, though the benefit of postoperative radiation therapy (RT) has not been established. In this study of grossly resected ACC, we compare local control of patients treated with surgery followed by adjuvant RT to a matched cohort treated with surgery alone. METHODS AND MATERIALS We retrospectively identified patients with localized disease who underwent R0 or R1 resection followed by adjuvant RT. Only patients treated with RT at our institution were included. Matching to surgical controls was on the basis of stage, surgical margin status, tumor grade, and adjuvant mitotane. RESULTS From 1991 to 2011, 360 ACC patients were evaluated for ACC at the University of Michigan (Ann Arbor, MI). Twenty patients with localized disease received postoperative adjuvant RT. These were matched to 20 controls. There were no statistically significant differences between the groups with regard to stage, margins, grade, or mitotane. Median RT dose was 55 Gy (range, 45-60 Gy). Median follow-up was 34 months. Local recurrence occurred in 1 patient treated with RT, compared with 12 patients not treated with RT (P=.0005; hazard ratio [HR] 12.59; 95% confidence interval [CI] 1.62-97.88). However, recurrence-free survival was no different between the groups (P=.17; HR 1.52; 95% CI 0.67-3.45). Overall survival was also not significantly different (P=.13; HR 1.97; 95% CI 0.57-6.77), with 4 deaths in the RT group compared with 9 in the control group. CONCLUSIONS Postoperative RT significantly improved local control compared with the use of surgery alone in this case-matched cohort analysis of grossly resected ACC patients. Although this retrospective series represents the largest study to date on adjuvant RT for ACC, its findings need to be prospectively confirmed.
Collapse
|
24
|
Guillaud-Bataille M, Ragazzon B, de Reyniès A, Chevalier C, Francillard I, Barreau O, Steunou V, Guillemot J, Tissier F, Rizk-Rabin M, René-Corail F, Ghuzlan AA, Assié G, Bertagna X, Baudin E, Le Bouc Y, Bertherat J, Clauser E. IGF2 promotes growth of adrenocortical carcinoma cells, but its overexpression does not modify phenotypic and molecular features of adrenocortical carcinoma. PLoS One 2014; 9:e103744. [PMID: 25089899 PMCID: PMC4121173 DOI: 10.1371/journal.pone.0103744] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/01/2014] [Indexed: 12/04/2022] Open
Abstract
Insulin-like growth factor 2 (IGF2) overexpression is an important molecular marker of adrenocortical carcinoma (ACC), which is a rare but devastating endocrine cancer. It is not clear whether IGF2 overexpression modifies the biology and growth of this cancer, thus more studies are required before IGF2 can be considered as a major therapeutic target. We compared the phenotypical, clinical, biological, and molecular characteristics of ACC with or without the overexpression of IGF2, to address these issues. We also carried out a similar analysis in an ACC cell line (H295R) in which IGF2 expression was knocked down with si- or shRNA. We found no significant differences in the clinical, biological and molecular (transcriptomic) traits between IGF2-high and IGF2-low ACC. The absence of IGF2 overexpression had little influence on the activation of tyrosine kinase pathways both in tumors and in H295 cells that express low levels of IGF2. In IGF2-low tumors, other growth factors (FGF9, PDGFA) are more expressed than in IGF2-high tumors, suggesting that they play a compensatory role in tumor progression. In addition, IGF2 knock-down in H295R cells substantially impaired growth (>50% inhibition), blocked cells in G1 phase, and promoted apoptosis (>2-fold). Finally, analysis of the 11p15 locus showed a paternal uniparental disomy in both IGF2-high and IGF2-low tumors, but low IGF2 expression could be explained in most IGF2-low ACC by an additional epigenetic modification at the 11p15 locus. Altogether, these observations confirm the active role of IGF2 in adrenocortical tumor growth, but also suggest that other growth promoting pathways may be involved in a subset of ACC with low IGF2 expression, which creates opportunities for the use of other targeted therapies.
Collapse
Affiliation(s)
- Marine Guillaud-Bataille
- Paris Cardiovascular Center, Institut National de la Santé et de la Recherche Médicale U970, Université Paris Descartes, Paris, France
- Département de Biologie Hormonale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Bruno Ragazzon
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Claire Chevalier
- Paris Cardiovascular Center, Institut National de la Santé et de la Recherche Médicale U970, Université Paris Descartes, Paris, France
| | - Isabelle Francillard
- Paris Cardiovascular Center, Institut National de la Santé et de la Recherche Médicale U970, Université Paris Descartes, Paris, France
- Département de Biologie Hormonale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Olivia Barreau
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
- Département d'Endocrinologie, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Virginie Steunou
- Institut National de la Santé et de la Recherche Médicale U938, Université Pierre et Marie Curie, Paris, France
- Laboratoire d'explorations fonctionnelles endocriniennes, Assistance Publique Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Johann Guillemot
- Paris Cardiovascular Center, Institut National de la Santé et de la Recherche Médicale U970, Université Paris Descartes, Paris, France
| | - Frédérique Tissier
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
- Service d'Anatomie Pathologique, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpétrière, Université Pierre et Marie Curie, Paris, France
| | - Marthe Rizk-Rabin
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
| | - Fernande René-Corail
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
| | - Abir Al Ghuzlan
- Département de Biologie et Pathologie Médicales, Institut Gustave Roussy, Villejuif, France
| | - Guillaume Assié
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
- Département d'Endocrinologie, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Xavier Bertagna
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
- Département d'Endocrinologie, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Eric Baudin
- Département d'Imagerie Médicale, Médecine nucléaire, Institut Gustave Roussy, Villejuif, France
| | - Yves Le Bouc
- Institut National de la Santé et de la Recherche Médicale U938, Université Pierre et Marie Curie, Paris, France
- Laboratoire d'explorations fonctionnelles endocriniennes, Assistance Publique Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Jérôme Bertherat
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Paris, France
- Département d'Endocrinologie, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Eric Clauser
- Paris Cardiovascular Center, Institut National de la Santé et de la Recherche Médicale U970, Université Paris Descartes, Paris, France
- Département de Biologie Hormonale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
- * E-mail:
| |
Collapse
|
25
|
Adrenocortical carcinoma: the management of metastatic disease. Crit Rev Oncol Hematol 2014; 92:123-32. [PMID: 24958272 DOI: 10.1016/j.critrevonc.2014.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/30/2014] [Accepted: 05/16/2014] [Indexed: 12/13/2022] Open
Abstract
Adrenocortical cancer is a rare malignancy. While surgery is the cornerstone of the management of localized disease, metastatic disease is hard to treat. Cytotoxic chemotherapy and mitotane have been utilized with a variable degree of benefit and few long-term responses. A growing understanding of the molecular pathogenesis of this malignancy as well as multidisciplinary and multi-institutional collaborative efforts will result in better defined targets and subsequently, effective novel therapies.
Collapse
|
26
|
Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano TJ, Hammer GD. Adrenocortical carcinoma. Endocr Rev 2014; 35:282-326. [PMID: 24423978 PMCID: PMC3963263 DOI: 10.1210/er.2013-1029] [Citation(s) in RCA: 596] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy, often with an unfavorable prognosis. Here we summarize the knowledge about diagnosis, epidemiology, pathophysiology, and therapy of ACC. Over recent years, multidisciplinary clinics have formed and the first international treatment trials have been conducted. This review focuses on evidence gained from recent basic science and clinical research and provides perspectives from the experience of a large multidisciplinary clinic dedicated to the care of patients with ACC.
Collapse
Affiliation(s)
- Tobias Else
- MEND/Division of Metabolism, Endocrinology, and Diabetes (T.E., T.J.G., G.D.H.), Division of Molecular Medicine and Genetics (V.M.R.), Department of Internal Medicine; Departments of Radiation Oncology (A.S., J.S.), Pathology (T.J.G.), and Radiology (A.K., E.M.C.); and Division of Endocrine Surgery (B.S.M.), Section of General Surgery, (A.C.K.), Department of Surgery, University of Michigan Hospital and Health Systems, Ann Arbor, Michigan 48109
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pilon C, Urbanet R, Williams TA, Maekawa T, Vettore S, Sirianni R, Pezzi V, Mulatero P, Fassina A, Sasano H, Fallo F. 1α,25-Dihydroxyvitamin D₃ inhibits the human H295R cell proliferation by cell cycle arrest: a model for a protective role of vitamin D receptor against adrenocortical cancer. J Steroid Biochem Mol Biol 2014; 140:26-33. [PMID: 24269839 DOI: 10.1016/j.jsbmb.2013.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/28/2013] [Accepted: 11/07/2013] [Indexed: 01/07/2023]
Abstract
UNLABELLED Using the human H295R adrenocortical carcinoma cell line as a model, we analyzed the role of 1α,25-dihydroxyvitamin D₃ [1α,25(OH)₂D₃)]--vitamin D receptor (VDR) axis in the growth of adrenocortical cancer (ACC). The presence of VDR in various adrenocortical tissues, including ACC, was also investigated. DNA synthesis was evaluated by [³H]thymidine cell incorporation after treatment with 1α,25(OH)₂D₃ at increasing doses. The effect of 1α,25(OH)₂D₃ on cell cycle and apoptosis was analyzed with a flow cytometer. Cyclin-dependent kinase 4 (CDK4) expression, a molecular marker of G1-S cell cycle transition phase, was evaluated in cells treated with 1α,25(OH)₂D₃ before and after VDR gene silencing. 1α,25(OH)₂D₃ treatment inhibited cell proliferation by 20% at a dose of 1 nM, in parallel with steroid secretion decrease. A cell cycle arrest in G1, with no change in apoptotic cell proportion, was observed after 10 nM 1α,25(OH)₂D₃ cell exposure. CDK4 activation was reduced by 10 nM 1α,25(OH)₂D₃ but was not affected by 1α,25(OH)₂D₃ after VDR gene silencing. Expression of VDR mRNA was lower in ACC than in benign adrenocortical tumors. VDR immunostaining was evident in benign tumors but it was weak in ACC tissues. CONCLUSIONS Slightly supra-physiological concentrations of 1α,25(OH)₂D₃ have a moderate anti-proliferative effect on H295R cells. Anti-proliferative effect was due to cell cycle arrest in G1 phase, without inducing apoptosis. The low mRNA expression levels at qRT-PCR as well as the weak immunohistochemical expression of VDR in ACC, suggests a protective role of VDR against malignant adrenocortical growth.
Collapse
Affiliation(s)
- Catia Pilon
- Department of Medicine-DIMED, Clinica Medica 3 and Cythopathology Unit, University of Padova, Padova, Italy
| | - Riccardo Urbanet
- Department of Medicine-DIMED, Clinica Medica 3 and Cythopathology Unit, University of Padova, Padova, Italy
| | - Tracy A Williams
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Takashi Maekawa
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | - Silvia Vettore
- Department of Medicine-DIMED, Clinica Medica 3 and Cythopathology Unit, University of Padova, Padova, Italy
| | - Rosa Sirianni
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria, Arcavacata di Rende (CS), Italy
| | - Paolo Mulatero
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Ambrogio Fassina
- Department of Medicine-DIMED, Clinica Medica 3 and Cythopathology Unit, University of Padova, Padova, Italy
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | - Francesco Fallo
- Department of Medicine-DIMED, Clinica Medica 3 and Cythopathology Unit, University of Padova, Padova, Italy.
| |
Collapse
|
28
|
Gentilin E, Molè D, Gagliano T, Minoia M, Ambrosio MR, Degli Uberti EC, Zatelli MC. Inhibitory effects of mitotane on viability and secretory activity in mouse gonadotroph cell lines. Reprod Toxicol 2014; 45:71-6. [PMID: 24486453 DOI: 10.1016/j.reprotox.2014.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/09/2014] [Accepted: 01/20/2014] [Indexed: 01/03/2023]
Abstract
Mitotane represents the mainstay medical treatment for metastatic, inoperable or recurrent adrenocortical carcinoma. Besides the well-known adverse events, mitotane therapy is associated also with endocrinological effects, including sexual and reproductive dysfunction. The majority of male patients undergoing adjuvant mitotane therapy show a picture of hypogonadism, characterized by low free testosterone and high sex hormone binding globulin levels and unmodified LH concentrations. Since mitotane has been shown to have direct pituitary effects, we investigated whether mitotane may influence both cell viability and function of gonadotroph cells in the settings of two pituitary cell lines. We found that mitotane reduces cell viability, induces apoptosis, modifies cell cycle phase distribution and secretion of gonadotroph cells. The present data strengthen previous evidence showing a direct mitotane effect at pituitary level and represent a possible explanation of the lack of LH increase following decrease in free testosterone in patients undergoing adjuvant mitotane therapy.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy
| | - Daniela Molè
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | - Teresa Gagliano
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | - Mariella Minoia
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | | | - Ettore C Degli Uberti
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy.
| |
Collapse
|
29
|
Germano A, Rapa I, Volante M, Lo Buono N, Carturan S, Berruti A, Terzolo M, Papotti M. Cytotoxic activity of gemcitabine, alone or in combination with mitotane, in adrenocortical carcinoma cell lines. Mol Cell Endocrinol 2014; 382:1-7. [PMID: 24018612 DOI: 10.1016/j.mce.2013.08.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 08/29/2013] [Accepted: 08/29/2013] [Indexed: 10/26/2022]
Abstract
We aimed at investigating in vitro the cytotoxic activity (determined using WST-1, apoptosis and cell cycle assays) of gemcitabine, alone or in combination with mitotane, in mitotane-sensitive H295R and mitotane-insensitive SW-13 cells. Results of these experiments were compared with drug-induced modulation of RRM1 gene, the specific target of gemcitabine. In H295R cells, mitotane and gemcitabine combinations showed antagonistic effects and interfered with the gemcitabine-mediated inhibition of the S phase of the cell cycle. By contrast, in SW-13 cells, except when mitotane was sequentially administered prior to gemcitabine, the combination of the two drugs was synergistic. Such opposite effects were associated with opposite expression profiles of the target gene, with significant up-modulation in H295R but not in SW-13 under gemcitabine and mitotane combination treatment.
Collapse
Affiliation(s)
- Antonina Germano
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Ida Rapa
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Marco Volante
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Nicola Lo Buono
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy.
| | - Sonia Carturan
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Alfredo Berruti
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Massimo Terzolo
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| | - Mauro Papotti
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, Turin, Italy.
| |
Collapse
|
30
|
Ho J, Turkbey B, Edgerly M, Alimchandani M, Quezado M, Camphausen K, Fojo T, Kaushal A. Role of radiotherapy in adrenocortical carcinoma. Cancer J 2013; 19:288-94. [PMID: 23867507 PMCID: PMC8381259 DOI: 10.1097/ppo.0b013e31829e3221] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Adrenocortical carcinoma (ACC) is a rare and highly malignant tumor usually diagnosed in an advanced stage. Radiation therapy has been a poorly studied and underutilized therapeutic option. METHODS This retrospective analysis reviewed treatment courses for 14 patients with pathologically confirmed ACC treated between 1997 and 2012. Two patients were treated adjuvantly following surgery, and 12 were treated with palliative intent. Patients presented with stage II (n = 4), stage III (n = 7), and stage IV (n = 3) disease. Patients had a mean age of 51.5 years. Ten patients received chemotherapy before radiotherapy (RT), and 12 patients received surgery before RT, before receiving radiation at a mean of 17.8 months after diagnosis. RESULTS In total, 20 sites were treated, 2 of which were in an adjuvant setting, and 18 of which were for palliative indications in 12 patients as follows: (1) pain/neuropathy (n = 10), (2) prophylactic treatment of asymptomatic recurrences (n = 3), and (3) prevention of imminent metastatic complications (n = 2), hemoptysis (n = 1), severe mass effect (n = 1), and brain metastasis (n = 1). Sites were treated to a median dose of 36.3 Gy (range, 17.5-60 Gy) in a median of 2.5 Gy/fraction (range, 1.8-4 Gy). At a mean follow-up of 22.0 months for the 2 patients given adjuvant RT, 1 patient did not have a local recurrence during a 14.3-month period of follow-up, and the other had a local recurrence 14.5 months after RT. At a mean follow-up of 11.3 months for the 12 patients receiving palliative RT, 10 patients had either a clinical or radiographic response. Of the courses of palliative RT that had adequate radiographic follow-up, 4 treatments (27%) resulted in a partial response. Eleven treatments (73%) that were able to be evaluated resulted in clinical improvement. Acute Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer toxicities observed in 7 patients included 3 grade 1, 4 grade 2, and 1 grade 3. No patient had acute toxicity of grade 4 or greater or any Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer late toxicity of grade 1 or greater. DISCUSSION This report is one of the largest to date examining the role of modern radiation techniques in the management of ACC. We conclude that radiation can be effective in the management of metastatic ACC, palliating local symptoms, and preventing complications from large metastases. Radiation should be considered as an option in multimodality management of ACC patients.
Collapse
Affiliation(s)
- Jennifer Ho
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maureen Edgerly
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Meghna Alimchandani
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tito Fojo
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Aradhana Kaushal
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
31
|
p53 Stabilization induces cell growth inhibition and affects IGF2 pathway in response to radiotherapy in adrenocortical cancer cells. PLoS One 2012; 7:e45129. [PMID: 23028800 PMCID: PMC3446967 DOI: 10.1371/journal.pone.0045129] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/14/2012] [Indexed: 01/28/2023] Open
Abstract
Adrenocortical carcinoma (ACC) is a very rare endocrine tumour, with variable prognosis, depending on tumour stage and time of diagnosis. However, it is generally fatal, with an overall survival of 5 years from detection. Radiotherapy usefulness for ACC treatment has been widely debated and seems to be dependent on molecular alterations, which in turn lead to increased radio-resistance. Many studies have shown that p53 loss is an important risk factor for malignant adrenocortical tumour onset and it has been reported that somatic mutations in TP53 gene occur in 27 to 70% of adult sporadic ACCs. In this study, we investigated the role of somatic mutations of the TP53 gene in response to ionizing radiation (IR). We studied the status of p53 in two adrenocortical cell lines, H295R and SW-13, harbouring non-functioning forms of this protein, owing to the lack of exons 8 and 9 and a point mutation in exon 6, respectively. Moreover, these cell lines show high levels of p-Akt and IGF2, especially H295R. We noticed that restoration of p53 activity led to inhibition of growth after transient transfection of cells with wild type p53. Evaluation of their response to IR in terms of cell proliferation and viability was determined by means of cell count and TUNEL assay.(wt)p53 over-expression also increased cell death by apoptosis following radiation in both cell lines. Moreover, RT-PCR and Western blotting analysis of some p53 target genes, such as BCL2, IGF2 and Akt demonstrated that p53 activation following IR led to a decrease in IGF2 expression. This was associated with a reduction in the active form of Akt. Taken together, these results highlight the role of p53 in response to radiation of ACC cell lines, suggesting its importance as a predictive factor for radiotherapy in malignant adrenocortical tumours cases.
Collapse
|
32
|
Zsippai A, Szabó DR, Tömböl Z, Szabó PM, Éder K, Pállinger É, Gaillard RC, Patócs A, Tóth S, Falus A, Rácz K, Igaz P. Effects of mitotane on gene expression in the adrenocortical cell line NCI-H295R: a microarray study. Pharmacogenomics 2012; 13:1351-61. [DOI: 10.2217/pgs.12.116] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The adrenolytic agent mitotane is widely used in the treatment of adrenocortical cancer; however, its mechanism of action is poorly elucidated. We have studied mitotane-induced mRNA expression changes in the NCI-H295R adrenocortical cancer cell line. Materials & methods: Cell viability and hormone assays were used to select the optimal mitotane concentration effectively inhibiting hormone secretion without affecting cell viability. RNA isolated from cultures treated for 48 and 72 h was subjected to Agilent 4×44K microarray platforms. Microarray results were validated by quantitative reverse-transcription PCR. Results: Altogether, 117 significantly differentially expressed genes were detected at 48 h and 72 h (p < 0.05) in mitotane-treated samples relative to controls. Three significantly underexpressed genes involved in steroid hormone biosynthesis (HSD3B1, HSD3B2 and CYP21A2) and four significantly overexpressed genes (GDF15, ALDH1L2, TRIB3 and SERPINE2) have been validated. Conclusion: Gene-expression changes might be involved in the adrenal action of mitotane and in the inhibition of hormone secretion. Original submitted 20 January 2012; Revision submitted 17 May 2012
Collapse
Affiliation(s)
- Adrienn Zsippai
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Diana Rita Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Zsófia Tömböl
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Peter M Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Katalin Éder
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Rolf C Gaillard
- Division of Endocrinology, Diabetology & Metabolism, University Hospital Lausanne, Rue du Bugnon 46., CH-1011 Lausanne, Switzerland
| | - Attila Patócs
- Molecular Medicine Research Group, Hungarian Academy of Sciences & Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Sára Tóth
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - András Falus
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Károly Rácz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Peter Igaz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| |
Collapse
|
33
|
Current and emerging therapeutic options in adrenocortical cancer treatment. JOURNAL OF ONCOLOGY 2012; 2012:408131. [PMID: 22934112 PMCID: PMC3425859 DOI: 10.1155/2012/408131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 12/14/2022]
Abstract
Adrenocortical carcinoma (ACC) is a very rare endocrine tumour, with variable prognosis, depending on tumour stage and time of diagnosis. The overall survival is five years from detection. Radical surgery is considered the therapy of choice in the first stages of ACC. However postoperative disease-free survival at 5 years is only around 30% and recurrence rates are frequent. o,p'DDD (ortho-, para'-, dichloro-, diphenyl-, dichloroethane, or mitotane), an adrenolytic drug with significant toxicity and unpredictable therapeutic response, is used in the treatment of ACC. Unfortunately, treatment for this aggressive cancer is still ineffective. Over the past years, the growing interest in ACC has contributed to the development of therapeutic strategies in order to contrast the neoplastic spread. In this paper we discuss the most promising therapies which can be used in this endocrine neoplasia.
Collapse
|
34
|
Milgrom SA, Goodman KA. The role of radiation therapy in the management of adrenal carcinoma and adrenal metastases. J Surg Oncol 2012; 106:647-50. [DOI: 10.1002/jso.23096] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/22/2012] [Indexed: 01/31/2023]
|
35
|
Nehs MA, Lin CI, Kozono DE, Whang EE, Cho NL, Zhu K, Moalem J, Moore FD, Ruan DT. Necroptosis is a novel mechanism of radiation-induced cell death in anaplastic thyroid and adrenocortical cancers. Surgery 2012; 150:1032-9. [PMID: 22136818 DOI: 10.1016/j.surg.2011.09.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 09/13/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Necroptosis is a recently described mechanism of programmed cellular death. We hypothesize that necroptosis plays an important role in radiation-induced cell death in endocrine cancers. METHODS Thyroid and adrenocortical carcinoma cell lines were exposed to increasing doses of radiation in the presence of necroptosis inhibitor Nec-1 and/or apoptosis-inhibitor zVAD. H295R cells deficient in receptor interacting protein 1 (RIP1), an essential kinase for necroptosis, were used as controls. Survival curves were generated at increasing doses of radiation. RESULTS Nec-1 and zVAD increased cellular survival with increasing doses of radiotherapy in 8505c, TPC-1, and SW13. Both inhibitors used together had an additive effect. At 6 Gy, 8505c, TPC-1, and SW13 cell survival was significantly increased compared to controls by 40%, 33%, and 31% with Nec-1 treatment, by 53%, 47%, and 44% with zVAD treatment, and by 80%, 70%, and 65% with both compounds, respectively (P < .05). H295R showed no change in survival with Nec-1 treatment. The radiobiologic parameter quasithreshold dose was significantly increased in 8505c, TPC-1, and SW13 cells when both Nec-1 and zVAD were used in combination to inhibit necroptosis and apoptosis together, revealing resistance to standard doses of fractionated therapeutic radiation. CONCLUSION Necroptosis contributes to radiation-induced cell death. Future studies should investigate ways to promote the activation of necroptosis to improve radiosensitivity.
Collapse
Affiliation(s)
- Matthew A Nehs
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tacon LJ, Prichard RS, Soon PSH, Robinson BG, Clifton-Bligh RJ, Sidhu SB. Current and emerging therapies for advanced adrenocortical carcinoma. Oncologist 2011; 16:36-48. [PMID: 21212436 DOI: 10.1634/theoncologist.2010-0270] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare but aggressive malignancy with a poor prognosis. Complete surgical resection offers the only potential for cure; however, even after apparently successful excision, local or metastatic recurrence is frequent. Treatment options for advanced ACC are severely limited. Mitotane is the only recognized adrenolytic therapy available; however, response rates are modest and unpredictable whereas systemic toxicities are significant. Reported responses to conventional cytotoxic chemotherapy have also been disappointing, and the rarity of ACC had hampered the ability to undertake randomized clinical studies until the establishment of the First International Randomized Trial in Locally Advanced and Metastatic Adrenocortical Carcinoma. This yet-to-be reported study seeks to identify the most effective first- and second-line cytotoxic regimens. The past decade has also seen increasing research into the molecular pathogenesis of ACCs, with particular interest in the insulin-like growth factor signaling pathway. The widespread development of small molecule tyrosine kinase inhibitors in broader oncological practice is now allowing for the rational selection of targeted therapies to study in ACC. In this review, we discuss the currently available therapeutic options for patients with advanced ACC and detail the molecular rationale behind, and clinical evidence for, novel and emerging therapies.
Collapse
Affiliation(s)
- Lyndal J Tacon
- Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research, Department of Endocrinology, Royal North Shore Hospital, St. Leonards 2065 NSW Australia.
| | | | | | | | | | | |
Collapse
|
37
|
Sabolch A, Feng M, Griffith K, Hammer G, Doherty G, Ben-Josef E. Adjuvant and definitive radiotherapy for adrenocortical carcinoma. Int J Radiat Oncol Biol Phys 2010; 80:1477-84. [PMID: 20675074 DOI: 10.1016/j.ijrobp.2010.04.030] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 03/08/2010] [Accepted: 04/23/2010] [Indexed: 01/14/2023]
Abstract
PURPOSE To evaluate the impact of both adjuvant and definitive radiotherapy on local control of adrenocortical carcinoma. METHODS AND MATERIALS Outcomes were analyzed from 58 patients with 64 instances of treatment for adrenocortical carcinoma at the University of Michigan's Multidisciplinary Adrenal Cancer Clinic. Thirty-seven of these instances were for primary disease, whereas the remaining 27 were for recurrent disease. Thirty-eight of the treatment regimens involved surgery alone, 10 surgery plus adjuvant radiotherapy, and 16 definitive radiotherapy for unresectable disease. The effects of patient, tumor, and treatment factors were modeled simultaneously using multiple variable Cox proportional hazards regression for associations with local recurrence, distant recurrence, and overall survival. RESULTS Local failure occurred in 16 of the 38 instances that involved surgery alone, in 2 of the 10 that consisted of surgery plus adjuvant radiotherapy, and in 1 instance of definitive radiotherapy. Lack of radiotherapy use was associated with 4.7 times the risk of local failure compared with treatment regimens that involved radiotherapy (95% confidence interval, 1.2-19.0; p = 0.030). CONCLUSIONS Radiotherapy seems to significantly lower the risk of local recurrence/progression in patients with adrenocortical carcinoma. Adjuvant radiotherapy should be strongly considered after surgical resection.
Collapse
Affiliation(s)
- Aaron Sabolch
- University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
38
|
Zatelli MC, Gentilin E, Daffara F, Tagliati F, Reimondo G, Carandina G, Ambrosio MR, Terzolo M, Degli Uberti EC. Therapeutic concentrations of mitotane (o,p'-DDD) inhibit thyrotroph cell viability and TSH expression and secretion in a mouse cell line model. Endocrinology 2010; 151:2453-61. [PMID: 20392828 DOI: 10.1210/en.2009-1404] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mitotane therapy is associated with many side effects, including thyroid function perturbations mimicking central hypothyroidism, possibly due to laboratory test interference or pituitary direct effects of mitotane. We investigated whether increasing concentrations of mitotane in the therapeutic range might interfere with thyroid hormone assays and evaluated the effects of mitotane on a mouse TSH-producing pituitary cell line. TSH, free T(4), and free T(3) levels do not significantly change in sera from hypo-, hyper-, or euthyroid patients after addition of mitotane at concentrations in the therapeutic window. In the mouse TalphaT1 cell line, mitotane inhibits both TSH expression and secretion, blocks TSH response to TRH, and reduces cell viability, inducing apoptosis at concentrations in the therapeutic window. TRH is not capable of rescuing TalphaT1 cells from the inhibitory effects of mitotane on TSH expression and secretion, which appear after short time treatment and persist over time. Our results demonstrate that mitotane does not interfere with thyroid hormone laboratory tests but directly reduces both secretory activity and cell viability on pituitary TSH-secreting mouse cells. These data represent a possible explanation of the biochemical picture consistent with central hypothyroidism in patients undergoing mitotane therapy and open new perspectives on the direct pituitary effects of this drug.
Collapse
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology, Department of Biomedical Sciences and Advanced Therapies, University of Ferrara, Via Savonarola 9, 44121 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Polat B, Fassnacht M, Pfreundner L, Guckenberger M, Bratengeier K, Johanssen S, Kenn W, Hahner S, Allolio B, Flentje M. Radiotherapy in adrenocortical carcinoma. Cancer 2009; 115:2816-23. [PMID: 19402169 DOI: 10.1002/cncr.24331] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy, and patients with ACC have a poor prognosis. Even after radical surgery, up to 85% of patients develop recurrent disease. Systemic treatment options still have limited efficacy. Because the role of radiotherapy is not defined well and because ACC often is considered radioresistant, the authors reviewed the available data on radiotherapy for ACC. Original articles and reviews were identified using a PubMed search strategy that included the period up to July 2008. Ten articles were identified that covered radiotherapy in a total of 129 patients with ACC (64 patients received postoperative irradiation, and 65 patients received palliative therapy for advanced disease). In addition, 26 patients were identified in the German ACC Registry who received palliative radiotherapy. Furthermore, patterns of failure after adjuvant radiotherapy were investigated, and the authors provided recommendations for patient selection, treatment planning, and treatment protocols. In an adjuvant setting, postoperative radiotherapy was able to prevent local recurrence in the majority of patients. In those with advanced disease, a response to radiotherapy was observed in 57% of patients who received palliative radiotherapy. Therefore, the authors concluded that radiotherapy may play an important role in the care of patients with ACC. Until better evidence is available, the authors recommended the following approach: Adjuvant radiotherapy to the tumor bed should be considered in patients at high risk for local recurrence (eg, incomplete/R1 resection); a total dose of >40 grays (Gy) with single fractions of 1.8 Gy to 2 Gy should be administered (including a boost volume to reach from 50 Gy to 60 Gy in individual patients); and radiotherapy in a palliative setting may be used for symptomatic metastases to bone, brain, or vena cava obstruction. With state-of-the-art technology, acute and long-term toxicities mostly were mild to moderate. However, the authors concluded that prospective investigations would be required to fully define the therapeutic potential of this important treatment option.
Collapse
Affiliation(s)
- Buelent Polat
- Department of Radiation Oncology, University Hospital, University of Wurzburg, Wurzburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|