1
|
Lalli E. Immune Checkpoint Molecules in Adrenocortical Carcinoma: Hope for Immunotherapy. J Clin Endocrinol Metab 2024; 110:e188-e189. [PMID: 38739542 DOI: 10.1210/clinem/dgae311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Affiliation(s)
- Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR7275, Inserm U1323, Université Côte d'Azur, 05650 Valbonne, France
| |
Collapse
|
2
|
Coral-Rivera NS, Vinasco A, Tascon-Barona AH. Adrenocortical neoplasm in a 2-year-old child: Clinical approach and diagnostic imaging. Radiol Case Rep 2024; 19:6417-6422. [PMID: 39380812 PMCID: PMC11460622 DOI: 10.1016/j.radcr.2024.09.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 10/10/2024] Open
Abstract
Adrenocortical tumors in children and adolescents are rare and aggressive, accounting for only 0.2% of pediatric cancers, with most cases associated with Li-Fraumeni syndrome. The most common manifestation is virilization due to androgen excess. Imaging techniques are crucial in the diagnosis and management of pediatric adrenocortical carcinoma. CT and MRI are essential for differentiating between benign and malignant lesions and assessing tumor characteristics and extent. Correlating imaging findings with clinical and histopathological data is vital for optimal diagnosis and treatment, underscoring the need for a multidisciplinary approach to managing these rare but aggressive neoplasms. This report presents the case of a previously healthy 2-year-old boy who exhibited virilization symptoms and was diagnosed with adrenocortical carcinoma.
Collapse
|
3
|
O'Neill AF, Ribeiro RC, Pinto EM, Clay MR, Zambetti GP, Orr BA, Weldon CB, Rodriguez-Galindo C. Pediatric Adrenocortical Carcinoma: The Nuts and Bolts of Diagnosis and Treatment and Avenues for Future Discovery. Cancer Manag Res 2024; 16:1141-1153. [PMID: 39263332 PMCID: PMC11389717 DOI: 10.2147/cmar.s348725] [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: 01/05/2024] [Accepted: 06/26/2024] [Indexed: 09/13/2024] Open
Abstract
Adrenocortical tumors (ACTs) are infrequent neoplasms in children and adolescents and are typically associated with clinical symptoms reflective of androgen overproduction. Pediatric ACTs typically occur in the context of a germline TP53 mutation, can be cured when diagnosed at an early stage, but are difficult to treat when advanced or associated with concurrent TP53 and ATRX alterations. Recent work has demonstrated DNA methylation patterns suggestive of prognostic significance. While current treatment standards rely heavily upon surgical resection, chemotherapy, and hormonal modulation, small cohort studies suggest promise for multi-tyrosine kinases targeting anti-angiogenic pathways or immunomodulatory therapies. Future work will focus on novel risk stratification algorithms and combination therapies intended to mitigate toxicity for patients with perceived low-risk disease while intensifying therapy or accelerating discoveries aimed at improving survival for patients with difficult-to-treat disease.
Collapse
Affiliation(s)
- Allison F O'Neill
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Raul C Ribeiro
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Emilia M Pinto
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael R Clay
- Department of Pathology, Children's Hospital Colorado, Denver, CO, USA
| | - Gerard P Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Carlos Rodriguez-Galindo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
4
|
Riedmeier M, Agarwal S, Antonini S, Costa TEIJB, Diclehan O, Fassnacht M, Figueiredo BC, Guran T, Härtel C, Haubitz I, Idkowiak J, Kuhlen M, Noronha L, Parise IZS, Redlich A, Puglisi S, Saniye E, Schlegel PG, Yalcin B, Wiegering V. Assessment of prognostic factors in pediatric adrenocortical tumors: the modified pediatric S-GRAS score in an international multicenter cohort-a work from the ENSAT-PACT working group. Eur J Endocrinol 2024; 191:64-74. [PMID: 38924056 DOI: 10.1093/ejendo/lvae079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Pediatric adrenocortical carcinoma (pACC) is rare, and prognostic stratification remains challenging. We aimed to confirm the prognostic value of the previously published pediatric scoring system (pS-GRAS) in an international multicenter cohort. DESIGN Analysis of pS-GRAS items of pACC from 6 countries in collaboration of ENSAT-PACT, GPOH-MET, and IC-PACT. METHODS We received patient data of the pS-GRAS items including survival information from 9 centers. PS-GRAS score was calculated as a sum of tumor stage (1 = 0; 2-3 = 1; 4 = 2 points), grade (Ki67 index: 0%-9% = 0; 10%-19% = 1; ≥20% = 2 points), resection status (R0 = 0; RX/R1/R2 = 1 point), age (<4 years = 0; ≥4 years = 1 point), and hormone production (androgen production = 0; glucocorticoid-/mixed-/no-hormone production = 1 point) generating 8 scores and 4 groups (1: 0-2, 2: 3-4, 3: 5, 4: 6-7). Primary endpoint was overall survival (OS). RESULTS We included 268 patients with median age of 4 years. The analysis of the pS-GRAS score showed a significantly favorable prognosis in patients with a lower scoring compared to higher scoring groups (5-year OS: Group 1 98%; group 2 87% [hazard ratio {HR} of death 3.6, 95% CI of HR 1.6-8.2]; group 3 43% [HR of death 2.8, 95% CI 1.9-4.4]; group 4: OS 18% [HR of death 2.1, 95% CI 1.7-2.7]). In the multivariable analysis, age (HR of death 3.5, 95% CI 1.8-7.0), resection status (HR of death 5.5, 95% CI 2.7-11.1), tumor stage (HR of death 1.9, 95% CI of HR 1.2-3.0), and Ki67 index (HR of death 1.7, 95% CI 1.2-2.4) remained strong independent outcome predictors. Especially infants < 4 years showed more often low-risk constellations with a better OS for all tumor stages. CONCLUSION In an international multicenter study, we confirmed that the pS-GRAS score is strongly associated with overall survival among patients with pACC. Age, resection status, stage, and Ki67 index are important parameters for risk stratification.
Collapse
Affiliation(s)
- Maria Riedmeier
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
- KIONET, The Phase I/II Pediatric Oncology Network Bavaria, 91054 Erlangen, Germany
| | - Shipra Agarwal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sonir Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo 14051-200, Brazill
| | - Tatiana E I Jaick B Costa
- Hospital Infantil Joana Gusmão, Department of Pediatrics, 152 Rui Barbosa St., Florianópolis, SC 88025-300, Brazil
| | - Orhan Diclehan
- Department of Pediatric Oncology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey
| | - Martin Fassnacht
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
- Department of Medicine, Division of Endocrinology and Diabetes, University of Wuerzbrug Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
| | - Bonald C Figueiredo
- Pelé Pequeno Príncipe Research Institute and Pequeno Príncipe Faculty, Silva Jardim Avenue, Água Verde, Curitiba, PR 80.250-200, Brazil
| | - Tulay Guran
- Department of Pediatric Endocrinology and Diabetes Istanbul, Marmara University School of Medicine, Istanbul 34722, Turkey
| | - Christoph Härtel
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
| | - Imme Haubitz
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
| | - Jan Idkowiak
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B152TT, United Kingdom
- Department of Endocrinology and Diabetes, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, Birmingham B46NH, United Kingdom
- Centre of Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Michaela Kuhlen
- KIONET, The Phase I/II Pediatric Oncology Network Bavaria, 91054 Erlangen, Germany
- Pediatrics and Adolescents Medicine, Faculty of Augsburg, University of Augsburg, 86156 Augsburg, Germany
| | - Lúcia Noronha
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, 181 General Carneiro, Alto da Glória, Curitiba, PR 80060-900, Brazil
| | - Ivy Zortéa S Parise
- Hospital Infantil Joana Gusmão, Department of Pediatrics, 152 Rui Barbosa St., Florianópolis, SC 88025-300, Brazil
| | - Antje Redlich
- KIONET, The Phase I/II Pediatric Oncology Network Bavaria, 91054 Erlangen, Germany
- Pediatric Oncology, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Soraya Puglisi
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, Orbassano 10043, Italy
| | - Ekinci Saniye
- Department of Pediatric Oncology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey
| | - Paul-Gerhardt Schlegel
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
- KIONET, The Phase I/II Pediatric Oncology Network Bavaria, 91054 Erlangen, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
| | - Bilgehan Yalcin
- Department of Pediatric Oncology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey
| | - Verena Wiegering
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
- KIONET, The Phase I/II Pediatric Oncology Network Bavaria, 91054 Erlangen, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany
- Mildred Scheel Early Career Center, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| |
Collapse
|
5
|
Riedmeier M, Antonini SRR, Brandalise S, Costa TEJB, Daiggi CM, de Figueiredo BC, de Krijger RR, De Sá Rodrigues KE, Deal C, Del Rivero J, Engstler G, Fassnacht M, Fernandes Luiz Canali GC, Molina CAF, Gonc EN, Gültekin M, Haak HR, Guran T, Hendriks Allaird EJ, Idkowiak J, Kuhlen M, Malkin D, Meena JP, Pamporaki C, Pinto E, Puglisi S, Ribeiro RC, Thompson LDR, Yalcin B, Van Noesel M, Wiegering V. International consensus on mitotane treatment in pediatric patients with adrenal cortical tumors: indications, therapy, and management of adverse effects. Eur J Endocrinol 2024; 190:G15-G24. [PMID: 38552173 DOI: 10.1093/ejendo/lvae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/17/2024] [Accepted: 02/19/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVE Mitotane is an important cornerstone in the treatment of pediatric adrenal cortical tumors (pACC), but experience with the drug in the pediatric age group is still limited and current practice is not guided by robust evidence. Therefore, we have compiled international consensus statements from pACC experts on mitotane indications, therapy, and management of adverse effects. METHODS A Delphi method with 3 rounds of questionnaires within the pACC expert consortium of the international network groups European Network for the Study of Adrenal Tumors pediatric working group (ENSAT-PACT) and International Consortium of pediatric adrenocortical tumors (ICPACT) was used to create 21 final consensus statements. RESULTS We divided the statements into 4 groups: environment, indications, therapy, and adverse effects. We reached a clear consensus for mitotane treatment for advanced pACC with stages III and IV and with incomplete resection/tumor spillage. For stage II patients, mitotane is not generally indicated. The timing of initiating mitotane therapy depends on the clinical condition of the patient and the setting of the planned therapy. We recommend a starting dose of 50 mg/kg/d (1500 mg/m²/d) which can be increased up to 4000 mg/m2/d. Blood levels should range between 14 and 20 mg/L. Duration of mitotane treatment depends on the clinical risk profile and tolerability. Mitotane treatment causes adrenal insufficiency in virtually all patients requiring glucocorticoid replacement shortly after beginning. As the spectrum of adverse effects of mitotane is wide-ranging and can be life-threatening, frequent clinical and neurological examinations (every 2-4 weeks), along with evaluation and assessment of laboratory values, are required. CONCLUSIONS The Delphi method enabled us to propose an expert consensus statement, which may guide clinicians, further adapted by local norms and the individual patient setting. In order to generate evidence, well-constructed studies should be the focus of future efforts.
Collapse
Affiliation(s)
- Maria Riedmeier
- University Hospital Würzburg, Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Wuerzburg, Wuerzburg 97080, Germany
| | - Sonir R R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo 14051-200, Brazil
| | - Silvia Brandalise
- Boldrini Children's Hospital, Department of Pediatrics, São Paulo 13083-210, Brazil
| | - Tatiana El Jaick B Costa
- Service of Pediatric Oncology, Hospital Infantil Joana de Gusmão, Florianópolis, SC 88025-301, Brazil
| | - Camila M Daiggi
- Boldrini Children's Hospital, Department of Pediatrics, São Paulo 13083-210, Brazil
| | | | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CS, The Netherlands
| | | | - Cheri Deal
- Research Center, CHU Sainte-Justine and University of Montreal, Montreal, Québec H3T 1C5, Canada
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Gernot Engstler
- St. Anna Kinderspital, Department of Pediatrics, Medical University Vienna, Vienna 1090, Austria
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg 97080, Germany
| | | | - Carlos A Fernandes Molina
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao 15 Paulo, Ribeirao Preto, Sao Paulo 14049-900, Brazil
| | - Elmas Nazli Gonc
- Department of Pediatric Endocrinology, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey
| | - Melis Gültekin
- Department of Radiation Oncology, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Harm R Haak
- Department of Internal Medicine, Máxima MC, Eindhoven 5631 BM/Veldhoven 5504 DB, The Netherlands
- CAPHRI School for Public Health and Primary Care, Ageing and Long-Term Care, Maastricht University, Maastricht 616 6200 MD, The Netherlands
| | - Tulay Guran
- Department of Paediatric Endocrinology and Diabetes, Marmara University School of Medicine, Istanbul 34722, Turkey
| | - Emile J Hendriks Allaird
- Department of Paediatrics, University of Cambridge, Cambridge CB20QQ, United Kingdom
- Department of Paediatric Endocrinology and Diabetes, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB20QQ, United Kingdom
| | - Jan Idkowiak
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B152TT, United Kingdom
- Department of Endocrinology, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham B46NH, United Kingdom
| | - Michaela Kuhlen
- Pediatrics and Adolescent Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86135, Germany
| | - David Malkin
- Division of Haematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G1X8, Canada
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, Mother & Child Block, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Christina Pamporaki
- Department of Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden 01307, Germany
| | - Emilia Pinto
- St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Soraya Puglisi
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano 10043, Italy
| | - Raul C Ribeiro
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Lester D R Thompson
- Pathology, Head and Neck Pathology Consultations, Woodland Hills, CA 91364, United States
| | - Bilgehan Yalcin
- Department of Pediatric Oncology, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey
| | - Max Van Noesel
- Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht 3584 CS, The Netherlands
- Division Imaging & Cancer, University Medical Center Utrecht, Utrecht 3584 CS, The Netherlands
| | - Verena Wiegering
- University Hospital Würzburg, Department of Pediatrics, Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Wuerzburg, Wuerzburg 97080, Germany
| |
Collapse
|
6
|
Zagojska E, Malka M, Gorecka A, Ben-Skowronek I. Case Report: Adrenocortical carcinoma in children-symptoms, diagnosis, and treatment. Front Endocrinol (Lausanne) 2023; 14:1216501. [PMID: 38075063 PMCID: PMC10702754 DOI: 10.3389/fendo.2023.1216501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Adrenocortical carcinomas are extremely rare in the paediatric population. Most of them are hormone-secretive lesions; therefore, they should be taken into consideration in a child with signs of precocious puberty and/or Cushing's syndrome symptoms. Nonetheless, differentiation from benign adrenal tumours is necessary. We report a rare case of adrenocortical carcinoma in a girl and a literature review using the PubMed database. A four-year-old girl presented with rapidly progressing precocious puberty and signs of Cushing's syndrome. Imaging of the abdomen revealed a large heterogeneous solid mass. Histopathologic evaluation confirmed adrenocortical carcinoma with high mitotic activity, atypical mitoses, pleomorphism, necrosis, and vascular invasion. After tumourectomy, a decrease of previously elevated hormonal blood parameters was observed. Genetic tests confirmed Li Fraumeni syndrome. Adrenocortical carcinoma should be suspected in children with premature pubarche and signs of Cushing's syndrome. Diagnosis must be based on clinical presentation, hormonal tests, imaging, and histopathological evaluation. Complete surgical resection of the tumour is the gold standard. Oncological treatment in children is not yet well-studied and should be individually considered, especially in advanced, inoperable carcinomas with metastases. Genetic investigations are useful for determining the prognosis in patients and their siblings.
Collapse
Affiliation(s)
| | | | | | - Iwona Ben-Skowronek
- Department of Paediatric Endocrinology and Diabetology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
7
|
The Characteristics of Tumor Microenvironment Predict Survival and Response to Immunotherapy in Adrenocortical Carcinomas. Cells 2023; 12:cells12050755. [PMID: 36899891 PMCID: PMC10000893 DOI: 10.3390/cells12050755] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Increasing evidence confirms that tumor microenvironment (TME) can influence tumor progression and treatment, but TME is still understudied in adrenocortical carcinoma (ACC). In this study, we first scored TME using the xCell algorithm, then defined genes associated with TME, and then used consensus unsupervised clustering analysis to construct TME-related subtypes. Meanwhile, weighted gene co-expression network analysis was used to identify modules correlated with TME-related subtypes. Ultimately, the LASSO-Cox approach was used to establish a TME-related signature. The results showed that TME-related scores in ACC may not correlate with clinical features but do promote a better overall survival. Patients were classified into two TME-related subtypes. Subtype 2 had more immune signaling features, higher expression of immune checkpoints and MHC molecules, no CTNNB1 mutations, higher infiltration of macrophages and endothelial cells, lower tumor immune dysfunction and exclusion scores, and higher immunophenoscore, suggesting that subtype 2 may be more sensitive to immunotherapy. 231 modular genes highly relevant to TME-related subtypes were identified, and a 7-gene TME-related signature that independently predicted patient prognosis was established. Our study revealed an integrated role of TME in ACC and helped to identify those patients who really responded to immunotherapy, while providing new strategies on risk management and prognosis prediction.
Collapse
|
8
|
Deng Y, Li H, Fu J, Pu Y, Zhang Y, Chen S, Tong S, Liu H. A hypoxia risk score for prognosis prediction and tumor microenvironment in adrenocortical carcinoma. Front Genet 2022; 13:796681. [PMID: 36583015 PMCID: PMC9792869 DOI: 10.3389/fgene.2022.796681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Adrenocortical carcinoma (ACC) is a rare malignant endocrine tumor derived from the adrenal cortex. Because of its highly aggressive nature, the prognosis of patients with adrenocortical carcinoma is not impressive. Hypoxia exists in the vast majority of solid tumors and contributes to invasion, metastasis, and drug resistance. This study aimed to reveal the role of hypoxia in Adrenocortical carcinoma and develop a hypoxia risk score (HRS) for Adrenocortical carcinoma prognostic prediction. Methods: Hypoxia-related genes were obtained from the Molecular Signatures Database. The training cohorts of patients with adrenocortical carcinoma were downloaded from The Cancer Genome Atlas, while another three validation cohorts with comprehensive survival data were collected from the Gene Expression Omnibus. In addition, we constructed a hypoxia classifier using a random survival forest model. Moreover, we explored the relationship between the hypoxia risk score and immunophenotype in adrenocortical carcinoma to evaluate the efficacy of immune check inhibitors (ICI) therapy and prognosis of patients. Results: HRS and tumor stage were identified as independent prognostic factors. HRS was negatively correlated with immune cycle activity, immune cell infiltration, and the T cell inflammatory score. Therefore, we considered the low hypoxia risk score group as the inflammatory immunophenotype, whereas the high HRS group was a non-inflammatory immunophenotype. In addition, the HRS was negatively related to the expression of common immune checkpoint molecules such as PD-L1, CD200, CTLA-4, and TIGIT, suggesting that patients with a lower hypoxia risk score respond better to immunotherapy. Conclusion: We developed and validated a novel hypoxia risk score to predict the immunophenotype and response of patients with adrenocortical carcinoma to immune check inhibitors therapy. These findings not only provide fresh prognostic indicators for adrenocortical carcinoma but also offer several promising treatment targets for this disease.
Collapse
Affiliation(s)
- Yuanyuan Deng
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinglan Fu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Pu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Zhang
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Shijing Chen
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Huixia Liu, ; Shiyu Tong,
| | - Huixia Liu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Huixia Liu, ; Shiyu Tong,
| |
Collapse
|
9
|
Wang Y, Zhang X, Xu C, Nan Y, Fan J, Zeng X, Kwon BS, Ju D. Targeting 4-1BB and PD-L1 induces potent and durable antitumor immunity in B-cell lymphoma. Front Immunol 2022; 13:1004475. [PMID: 36544785 PMCID: PMC9762552 DOI: 10.3389/fimmu.2022.1004475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Although PD-1/L1 mAb has demonstrated clinical benefits in certain cancer types, low response rate and resistance remain the main challenges for the application of these immune checkpoint inhibitors (ICIs). 4-1BB is a co-stimulator molecule expressed in T cells, which could enhance T cell proliferation and activation. Herein, the synergetic antitumor effect and underlying mechanism of 4-1BB agonist combined with PD-1/PD-L1 blockade were determined in B-cell lymphoma (BCL). Methods Subcutaneous transplantation BCL tumor models and metastasis models were established to evaluate the therapeutic effect of PD-L1 antibody and/or 4-1BB agonist in vivo. For the mechanistic study, RNA-seq was applied to analyze the tumor microenvironment and immune-related signal pathway after combination treatment. The level of IFN-γ, perforin, and granzyme B were determined by ELISA and Real-time PCR assays, while tumor-infiltrating T cells were measured by flow cytometry and immunohistochemical analysis. CD4/CD8 specific antibodies were employed to deplete the related T cells to investigate the role CD4+ and CD8+ T cells played in combination treatment. Results Our results showed that combining anti-PD-L1 ICI and 4-1BB agonists elicited regression of BCL and significantly extended the survival of mice compared to either monotherapy. Co-targeting PD-L1 and 4-1BB preferentially promoted intratumoral cytotoxic lymphocyte infiltration and remodeled their function. RNA-sequence analysis uncovered a series of up-regulated genes related to the activation and proliferation of cytotoxic T lymphocytes, further characterized by increased cytokines including IFN-γ, granzyme B, and perforin. Furthermore, depleting CD8+ T cells not CD4+ T cells totally abrogated the antitumor efficacy, indicating the crucial function of the CD8+ T cell subset in the combination therapy. Discussion In summary, our findings demonstrated that 4-1BB agonistic antibody intensified the antitumor immunity of anti-PD-1/PD-L1 ICI via promoting CD8+ T cell infiltration and activation, providing a novel therapeutic strategy to BCL.
Collapse
Affiliation(s)
- Yichen Wang
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Xuyao Zhang
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Caili Xu
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Yanyang Nan
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Jiajun Fan
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Xian Zeng
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| | - Byoung S. Kwon
- Eutilex Institute for Biomedical Research, Eutilex Co., Ltd, Seoul, South Korea
| | - Dianwen Ju
- School of Pharmacy and Minhang Hospital, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China,Department of Biologics, Fudan Zhangjiang Institute, Shanghai, China,*Correspondence: Dianwen Ju,
| |
Collapse
|
10
|
Riedmeier M, Decarolis B, Haubitz I, Reibetanz J, Wiegering A, Härtel C, Schlegel PG, Fassnacht M, Wiegering V. Assessment of prognostic factors in pediatric adrenocortical tumors: a systematic review and evaluation of a modified S-GRAS score. Eur J Endocrinol 2022; 187:751-763. [PMID: 36193775 DOI: 10.1530/eje-22-0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/03/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Pediatric adrenocortical carcinoma (pACC) is rare and prognostic stratification remains challenging. We summarized the clinical prognostic factors of pACC and determined the prognostic value of the pediatric scoring system (pS-GRAS) in adaption to the recommendation (S-GRAS) of the European Network for the Study of Adrenal Tumors for the classification of adult ACC. DESIGN Analysis of pACC patients of 33 available retrospective studies in the literature. METHODS We searched the PubMed and Embase databases for manuscripts regarding pACC. The pS-GRAS score was calculated as a sum of tumor stage (1 = 0; 2-3 = 1; 4 = 2 points), grade (Ki67 index/rate of mitosis 0-9%/low = 0; 10-19%/intermediate = 1; ≥20%/high = 2 points), resection status (R0 = 0; RX = 1; R1 = 2; R2 = 3 points), age (<4 years = 0; ≥4 years = 1 point), hormone-related symptoms (androgen production = 0; glucocorticoid/mixed/no hormone production = 1 point) generating 10 scores and 4 groups (1: 0-2, 2: 3-4, 3: 5, 4: 6-9). The primary endpoint was overall survival (OS). RESULTS We included 733 patients. The median age was 2.5 years and >85% of pACC showed hormone activity (mixed 50%, androgen 29%, glucocorticoid 21%). Androgen production was associated with a superior OS. Increasing age correlated with higher rates of inactive or only glucocorticoid-producing tumors, advanced tumor stage, and case fatality. Especially infants < 4 years showed more often low-risk constellations with an increased OS for all tumor stages. The pS-GRAS score correlated with clinical outcome; median OS was 133 months (95% CI: 36-283) in group 1 (n = 49), 110 months (95% CI: 2.9-314) in group 2 (n = 57), 49 months (95% CI: 5.8-278) in group 3 (n = 18), and 16 months (95% CI: 2.4-267) in group 4; (n = 11) P < 0.05). CONCLUSION The pS-GRAS score seems to have a high predictive value in the pACC patients, may serve as a helpful tool for risk stratification in future studies, and should be evaluated prospectively in an international context.
Collapse
Affiliation(s)
- Maria Riedmeier
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Medical Faculty, Cologne, Germany
| | - Imme Haubitz
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Joachim Reibetanz
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Armin Wiegering
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Wuerzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Wuerzburg, Germany
| | - Christoph Härtel
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University Hospital, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Wuerzburg, Germany
| | - Paul-Gerhardt Schlegel
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University Hospital, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Wuerzburg, Germany
| | - Martin Fassnacht
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Wuerzburg, Germany
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Verena Wiegering
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University Hospital, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Wuerzburg, Germany
| |
Collapse
|
11
|
Yu QX, Feng DC, Li DX, Zheng HH. Prognostic value of spindle and kinetochore-related complex family and its correlation with immune infiltration in adrenocortical carcinoma. Asian J Surg 2022; 46:1990-1992. [PMID: 36418213 DOI: 10.1016/j.asjsur.2022.10.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Qing-Xin Yu
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang Province, 317000, China
| | - De-Chao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Deng-Xiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Hai-Hong Zheng
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang Province, 317000, China.
| |
Collapse
|
12
|
Sandru F, Petca RC, Carsote M, Petca A, Dumitrascu M, Ghemigian A. Adrenocortical carcinoma: Pediatric aspects (Review). Exp Ther Med 2022; 23:287. [DOI: 10.3892/etm.2022.11216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Florica Sandru
- Department of Dermatology, ‘Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Răzvan-Cosmin Petca
- Department of Urology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mara Carsote
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihai Dumitrascu
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adina Ghemigian
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
13
|
Tosin KCF, Legal EF, Pianovski MAD, Ibañez HC, Custódio G, Carvalho DS, Figueiredo MMO, Hoffmann Filho A, Fiori CMCM, Rodrigues ALM, Mello RG, Ogradowski KRP, Parise IZS, Costa TEJ, Melanda VS, Watanabe FM, Silva DB, Komechen H, Laureano HA, Carboni EK, Kuczynski AP, Luiz GCF, Lima L, Tormen T, Gerber VKQ, Anegawa TH, Avilla SGA, Tenório RB, Mendes EL, Fachin Donin RD, Souza J, Kozak VN, Oliveira GS, Souza DC, Gomy I, Teixeira VB, Borba HHL, Kiesel Filho N, Parise GA, Ribeiro RC, Figueiredo BC. Newborn Screening for the Detection of the TP53 R337H Variant and Surveillance for Early Diagnosis of Pediatric Adrenocortical Tumors: Lessons Learned and Way Forward. Cancers (Basel) 2021; 13:cancers13236111. [PMID: 34885220 PMCID: PMC8656743 DOI: 10.3390/cancers13236111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Adrenocortical tumor (ACT) is rare in children and fatal if not detected early. Children who inherit a mutation of the TP53 gene tend to develop ACT early in life. In the 1990s, scientists revealed that a TP53 variant (R337H) was frequent in South Brazil. Therefore, the incidence of ACT in children is 20 times higher in this region than in other countries. We reviewed the records of 16 children with ACT treated in a pediatric hospital in Parana state (southern Brazil) and 134 children registered in the state public registry data. We found a high number of cases with advanced disease, leading to an unacceptable number of deaths. These observations contradict newborn R337H screening and surveillance data, showing that surgical intervention in early cases of ACT is associated with a 100% cure. Newborn screening/surveillance should be implemented in regions with a high frequency of the R337H variant. Abstract The incidence of pediatric adrenocortical tumors (ACT) is high in southern Brazil due to the founder TP53 R337H variant. Neonatal screening/surveillance (NSS) for this variant resulted in early ACT detection and improved outcomes. The medical records of children with ACT who did not participate in newborn screening (non-NSS) were reviewed (2012–2018). We compared known prognostic factors between the NSS and non-NSS cohorts and estimated surveillance and treatment costs. Of the 16 non-NSS children with ACT carrying the R337H variant, the disease stages I, II, III, and IV were observed in five, five, one, and five children, respectively. The tumor weight ranged from 22 to 608 g. The 11 NSS children with ACT all had disease stage I and were alive. The median tumor weight, age of diagnosis, and interval between symptoms and diagnosis were 21 g, 1.9 years, and two weeks, respectively, for the NSS cohort and 210 g, 5.2 years, and 15 weeks, respectively, for the non-NSS cohort. The estimated surveillance/screening cost per year of life saved is US$623/patient. NSS is critical for improving the outcome of pediatric ACT in this region. Hence, we strongly advocate for the inclusion of R337H in the state-mandated universal screening and surveillance.
Collapse
Affiliation(s)
- Karina C. F. Tosin
- Departamento de Saúde Coletiva, Federal University of Paraná, Rua Padre Camargo, 260, Centro, Curitiba 80.060-240, PR, Brazil; (K.C.F.T.); (D.S.C.)
| | - Edith F. Legal
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Mara A. D. Pianovski
- Oncologia Pediátrica, Hospital Erasto Gaertner, R. Dr. Ovande do Amaral, 201, Jardim das Américas, Curitiba 81.520-060, PR, Brazil; (M.A.D.P.); (A.L.M.R.); (V.N.K.); (G.S.O.); (D.C.S.)
| | - Humberto C. Ibañez
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Gislaine Custódio
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
| | - Denise S. Carvalho
- Departamento de Saúde Coletiva, Federal University of Paraná, Rua Padre Camargo, 260, Centro, Curitiba 80.060-240, PR, Brazil; (K.C.F.T.); (D.S.C.)
| | - Mirna M. O. Figueiredo
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
| | - Anselmo Hoffmann Filho
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Carmem M. C. M. Fiori
- Hospital do Câncer, UOPECCAN, R. Itaquatiaras, 769, Santo Onofre, Cascavel 85.806-300, PR, Brazil;
| | - Ana Luiza M. Rodrigues
- Oncologia Pediátrica, Hospital Erasto Gaertner, R. Dr. Ovande do Amaral, 201, Jardim das Américas, Curitiba 81.520-060, PR, Brazil; (M.A.D.P.); (A.L.M.R.); (V.N.K.); (G.S.O.); (D.C.S.)
| | - Rosiane G. Mello
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
- Faculdades Pequeno Príncipe, Av. Iguaçu, 333, Rebouças, Curitiba 80.230-020, PR, Brazil;
| | - Karin R. P. Ogradowski
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
- Faculdades Pequeno Príncipe, Av. Iguaçu, 333, Rebouças, Curitiba 80.230-020, PR, Brazil;
| | - Ivy Z. S. Parise
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Tatiana E. J. Costa
- Hospital Infantil Joana de Gusmão, R. Rui Barbosa, 152, Agronômica, Florianópolis 88.025-301, SC, Brazil; (T.E.J.C.); (D.B.S.)
| | - Viviane S. Melanda
- Secretaria do Estado da Saúde do Paraná, R. Piquiri, 170, Rebouças, Curitiba 80.230-140, PR, Brazil;
| | - Flora M. Watanabe
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Denise B. Silva
- Hospital Infantil Joana de Gusmão, R. Rui Barbosa, 152, Agronômica, Florianópolis 88.025-301, SC, Brazil; (T.E.J.C.); (D.B.S.)
| | - Heloisa Komechen
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
| | - Henrique A. Laureano
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Edna K. Carboni
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Ana P. Kuczynski
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Gabriela C. F. Luiz
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Leniza Lima
- Oncologia Pediátrica, Hospital de Clínicas da Universidade Federal do Paraná, R. Gen. Carneiro, 181, Alto da Glória, Curitiba 80.060-900, PR, Brazil; (L.L.); (T.T.)
| | - Tiago Tormen
- Oncologia Pediátrica, Hospital de Clínicas da Universidade Federal do Paraná, R. Gen. Carneiro, 181, Alto da Glória, Curitiba 80.060-900, PR, Brazil; (L.L.); (T.T.)
| | - Viviane K. Q. Gerber
- Departamento de Enfermagem, Universidade Estadual do Centro-Oeste, UNICENTRO, Rua Padre, R. Salvatore Renna, 875-Santa Cruz, Guarapuava 85.015-430, PR, Brazil;
| | - Tania H. Anegawa
- Oncologia Pediátrica, Campus Universitário, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid—Pr 445 Km 380, Londrina 86.057-970, PR, Brazil;
| | - Sylvio G. A. Avilla
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Renata B. Tenório
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Elaine L. Mendes
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Rayssa D. Fachin Donin
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
| | - Josiane Souza
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Vanessa N. Kozak
- Oncologia Pediátrica, Hospital Erasto Gaertner, R. Dr. Ovande do Amaral, 201, Jardim das Américas, Curitiba 81.520-060, PR, Brazil; (M.A.D.P.); (A.L.M.R.); (V.N.K.); (G.S.O.); (D.C.S.)
| | - Gisele S. Oliveira
- Oncologia Pediátrica, Hospital Erasto Gaertner, R. Dr. Ovande do Amaral, 201, Jardim das Américas, Curitiba 81.520-060, PR, Brazil; (M.A.D.P.); (A.L.M.R.); (V.N.K.); (G.S.O.); (D.C.S.)
| | - Deivid C. Souza
- Oncologia Pediátrica, Hospital Erasto Gaertner, R. Dr. Ovande do Amaral, 201, Jardim das Américas, Curitiba 81.520-060, PR, Brazil; (M.A.D.P.); (A.L.M.R.); (V.N.K.); (G.S.O.); (D.C.S.)
| | - Israel Gomy
- Faculdades Pequeno Príncipe, Av. Iguaçu, 333, Rebouças, Curitiba 80.230-020, PR, Brazil;
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Vinicius B. Teixeira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
| | - Helena H. L. Borba
- Departamento de Ciências Farmacêuticas, Federal University of Paraná, Av. Prefeito Lothário Meissner, 632-Jardim Botanico, Curitiba 80.210-170, PR, Brazil;
| | - Nilton Kiesel Filho
- Hospital Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (F.M.W.); (E.K.C.); (A.P.K.); (G.C.F.L.); (S.G.A.A.); (R.B.T.); (E.L.M.); (J.S.); (N.K.F.)
| | - Guilherme A. Parise
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
| | - Raul C. Ribeiro
- Leukemia and Lymphoma Division, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Correspondence: (R.C.R.); or (B.C.F.)
| | - Bonald C. Figueiredo
- Departamento de Saúde Coletiva, Federal University of Paraná, Rua Padre Camargo, 260, Centro, Curitiba 80.060-240, PR, Brazil; (K.C.F.T.); (D.S.C.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Silva Jardim, 1532, Curitiba 80.250-060, PR, Brazil; (E.F.L.); (H.C.I.); (A.H.F.); (R.G.M.); (K.R.P.O.); (I.Z.S.P.); (H.K.); (H.A.L.); (V.B.T.)
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC-APACN), Avenida Agostinho Leão Jr., 400, Curitiba 80.030-110, PR, Brazil; (G.C.); (M.M.O.F.); (R.D.F.D.); (G.A.P.)
- Faculdades Pequeno Príncipe, Av. Iguaçu, 333, Rebouças, Curitiba 80.230-020, PR, Brazil;
- Correspondence: (R.C.R.); or (B.C.F.)
| |
Collapse
|
14
|
Riedmeier M, Decarolis B, Haubitz I, Müller S, Uttinger K, Börner K, Reibetanz J, Wiegering A, Härtel C, Schlegel PG, Fassnacht M, Wiegering V. Adrenocortical Carcinoma in Childhood: A Systematic Review. Cancers (Basel) 2021; 13:5266. [PMID: 34771430 PMCID: PMC8582500 DOI: 10.3390/cancers13215266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/01/2023] Open
Abstract
Adrenocortical tumors are rare in children. This systematic review summarizes the published evidence on pediatric adrenocortical carcinoma (ACC) to provide a basis for a better understanding of the disease, investigate new molecular biomarkers and therapeutic targets, and define which patients may benefit from a more aggressive therapeutic approach. We included 137 studies with 3680 ACC patients (~65% female) in our analysis. We found no randomized controlled trials, so this review mainly reflects retrospective data. Due to a specific mutation in the TP53 gene in ~80% of Brazilian patients, that cohort was analyzed separately from series from other countries. Hormone analysis was described in 2569 of the 2874 patients (89%). Most patients were diagnosed with localized disease, whereas 23% had metastasis at primary diagnosis. Only 72% of the patients achieved complete resection. In 334 children (23%), recurrent disease was reported: 81%-local recurrence, 19% (n = 65)-distant metastases at relapse. Patients < 4 years old had a different distribution of tumor stages and hormone activity and better overall survival (p < 0.001). Although therapeutic approaches are typically multimodal, no consensus is available on effective standard treatments for advanced ACC. Thus, knowledge regarding pediatric ACC is still scarce and international prospective studies are needed to implement standardized clinical stratifications and risk-adapted therapeutic strategies.
Collapse
Affiliation(s)
- Maria Riedmeier
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, Medical Faculty, University Children’s Hospital of Cologne, 50937 Cologne, Germany;
| | - Imme Haubitz
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
| | - Sophie Müller
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Konstantin Uttinger
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Kevin Börner
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Joachim Reibetanz
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Armin Wiegering
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Christoph Härtel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Martin Fassnacht
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany
| | - Verena Wiegering
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| |
Collapse
|
15
|
Georgantzoglou N, Kokkali S, Tsourouflis G, Theocharis S. Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success? Cancers (Basel) 2021; 13:1798. [PMID: 33918733 PMCID: PMC8069982 DOI: 10.3390/cancers13081798] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Adrenocortical carcinoma is a rare malignancy with aggressive behavior, with up to 40% of patients presenting with metastases at the time of diagnosis. Both conventional chemotherapeutic regimens and novel immunotherapeutic agents, many of which are currently being tested in ongoing clinical trials, have yielded modest results so far, bringing the need for a deeper understanding of adrenal cancer behavior to the forefront. In the recent years, the tumor microenvironment has emerged as a major determinant of cancer response to immunotherapy and an increasing number of studies on other solid tumors have focused on manipulating the microenvironment in the favor of the host and discovering new potential target molecules. In the present review we aim to explore the characteristics of adrenocortical cancer's microenvironment, highlighting the mechanisms of immune evasion responsible for the modest immunotherapeutic results, and identify novel potential strategies.
Collapse
Affiliation(s)
- Natalia Georgantzoglou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (N.G.); (S.K.)
| | - Stefania Kokkali
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (N.G.); (S.K.)
- First Medical Oncology Clinic, Saint-Savvas Anti Cancer Hospital, 115 27 Athens, Greece
| | - Gerasimos Tsourouflis
- Second Department of Propedeutic Surgery, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (N.G.); (S.K.)
| |
Collapse
|
16
|
Landwehr LS, Altieri B, Schreiner J, Sbiera I, Weigand I, Kroiss M, Fassnacht M, Sbiera S. Interplay between glucocorticoids and tumor-infiltrating lymphocytes on the prognosis of adrenocortical carcinoma. J Immunother Cancer 2021; 8:jitc-2019-000469. [PMID: 32474412 PMCID: PMC7264832 DOI: 10.1136/jitc-2019-000469] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background Adrenocortical carcinoma (ACC) is a rare endocrine malignancy. Tumor-related glucocorticoid excess is present in ~60% of patients and associated with particularly poor prognosis. Results of first clinical trials using immune checkpoint inhibitors were heterogeneous. Here we characterize tumor-infiltrating T lymphocytes (TILs) in ACC in association with glucocorticoids as potential explanation for resistance to immunotherapy. Methods We performed immunofluorescence analysis to visualize tumor-infiltrating T cells (CD3+), T helper cells (CD3+CD4+), cytotoxic T cells (CD3+CD8+) and regulatory T cells (Tregs; CD3+CD4+FoxP3+) in 146 ACC tissue specimens (107 primary tumors, 16 local recurrences, 23 metastases). Quantitative data of immune cell infiltration were correlated with clinical data (including glucocorticoid excess). Results 86.3% of ACC specimens showed tumor infiltrating T cells (7.7 cells/high power field (HPF)), including T helper (74.0%, 6.7 cells/HPF), cytotoxic T cells (84.3%, 5.7 cells/HPF) and Tregs (49.3%, 0.8 cells/HPF). The number of TILs was associated with better overall survival (HR for death: 0.47, 95% CI 0.25 to 0.87), which was true for CD4+− and CD8+ subpopulations as well. In localized, non-metastatic ACC, the favorable impact of TILs on overall and recurrence-free survival was manifested even independently of ENSAT (European Network for the Study of Adrenal Tumors) stage, resection status and Ki67 index. T helper cells were negatively correlated with glucocorticoid excess (Phi=−0.290, p=0.009). Patients with glucocorticoid excess and low TILs had a particularly poor overall survival (27 vs. 121 months in patients with TILs without glucocorticoid excess). Conclusion Glucocorticoid excess is associated with T cell depletion and unfavorable prognosis. To reactivate the immune system in ACC by checkpoint inhibitors, an inhibition of adrenal steroidogenesis might be pivotal and should be tested in prospective studies.
Collapse
Affiliation(s)
- Laura-Sophie Landwehr
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Barbara Altieri
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Jochen Schreiner
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Iuliu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Isabel Weigand
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Matthias Kroiss
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany .,Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Silviu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany .,Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Berruti A, Tiberio GAM, Sigala S. Adrenocortical Carcinoma. Cancers (Basel) 2021; 13:1077. [PMID: 33802418 PMCID: PMC7959308 DOI: 10.3390/cancers13051077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 12/28/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is an extremely rare disease, the incidence of which is 0 [...].
Collapse
Affiliation(s)
- Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST Spedali Civili of Brescia, 25123 Brescia, Italy;
| | - Guido Alberto Massimo Tiberio
- Surgical Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST Spedali Civili of Brescia, 25123 Brescia, Italy;
| | - Sandra Sigala
- Department of Molecular & Translational Medicine, Section of Pharmacology, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
18
|
Zambaiti E, Duci M, De Corti F, Gamba P, Dall'Igna P, Ghidini F, Virgone C. Clinical prognostic factors in pediatric adrenocortical tumors: A meta-analysis. Pediatr Blood Cancer 2021; 68:e28836. [PMID: 33306282 DOI: 10.1002/pbc.28836] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/27/2020] [Accepted: 11/14/2020] [Indexed: 12/14/2022]
Abstract
Pediatric adrenocortical tumors (ACT) are rare and sometimes aggressive malignancies, but there is no consensus on the outcome predictors in children. A systematic search of MEDLINE, SCOPUS, Web of Science, and the Cochrane Library for studies from 1994 to 2020 about pediatric ACT was performed. In 42 studies, 1006 patients, aged 0-18 years, were included. The meta-analyses resulted in the following predictors of better outcome: age <4 years (P < .00001), nonsecreting tumors (P = .004), complete surgical resection (P < .00001), tumor volume (P < .0001), tumor weight (P < .00001), tumor maximum diameter (P = .0009), and Stage I disease (P < .00001). Moreover, patients affected by Cushing syndrome showed a worse outcome (P < .0001). International prospective studies should be implemented to standardize clinical prognostic factors evaluation, together with pathological scores, in the stratification of pediatric ACT.
Collapse
Affiliation(s)
- Elisa Zambaiti
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Miriam Duci
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Federica De Corti
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Piergiorgio Gamba
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Patrizia Dall'Igna
- Department of Emergencies and Organ Transplantation, Azienda Ospedaliero-Universitaria Consorziale Ospedale Pediatrico Giovanni XXIII, Bari, Italy
| | - Filippo Ghidini
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Calogero Virgone
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| |
Collapse
|
19
|
Muzzi JCD, Magno JM, Cardoso MA, de Moura J, Castro MAA, Figueiredo BC. Adrenocortical Carcinoma Steroid Profiles: In Silico Pan-Cancer Analysis of TCGA Data Uncovers Immunotherapy Targets for Potential Improved Outcomes. Front Endocrinol (Lausanne) 2021; 12:672319. [PMID: 34194394 PMCID: PMC8237859 DOI: 10.3389/fendo.2021.672319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Despite progress in understanding the biology of adrenocortical carcinoma (ACC), treatment options have not dramatically changed in the last three decades, nor have we learned how to avoid some of its long-term side effects. Our goal was to improve the understanding of immune pathways that may include druggable targets to enhance immune responses of patients with ACC, focusing on immune evasion and the activation of immune cells against ACC. Our strategy was aimed at improving insight regarding gene expression without steroid interference. Using approaches based on high and low steroid phenotypes (HSP and LSP, respectively), we characterized immune pathways using The Cancer Genome Atlas (TCGA) ACC cohort data. Although previous studies have suggested that patients with ACC receive minimal benefit from immunotherapy, high expression of immune modulators was noted in patients with LSP, suggesting the activation of these biomarkers may be an important adjuvant therapy target after clearance of excess glucocorticoids. In addition, patients with LSP ACC had higher immune cell infiltration than patients with HSP ACC and other cancer subtypes. Our findings can be summarized as follows (1): we confirmed and improved the definition of two immune response pathways to ACC (HSP and LSP) based on in silico transcriptome analysis (2), we demonstrated the steroid profile should be considered, otherwise analyses of ACC immune characteristics can generate confounding results (3), among the overexpressed immunotherapy targets, we demonstrated that LSP was rich in PDCD1LG2 (PD-L2) and both HSP and LSP overexpressed CD276 (B7-H3), which was associated with resistance to anti-PD1 therapy and may have accounted for the modest results of previous clinical trials, and (4) identification of patients with LSP or HSP ACC can be used to help determine whether immunotherapy should be used. In conclusion, we highlighted the differences between LSP and HSP, drawing attention to potential therapeutic targets (CD276, PDCD1, and PDCD1LG2). Treatments to reduce immune evasion, as well as the use of other natural and pharmacological immune activators, should include prior pharmacological inhibition of steroidogenesis. Attempts to combine these with tumor cell proliferation inhibitors, if they do not affect cells of the immune system, may produce interesting results.
Collapse
Affiliation(s)
- João C. D. Muzzi
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba, Brazil
| | - Jessica M. Magno
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba, Brazil
| | - Milena A. Cardoso
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba, Brazil
| | - Juliana de Moura
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Mauro A. A. Castro
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Bonald C. Figueiredo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Molecular Oncology Laboratory, Curitiba, Brazil
- *Correspondence: Bonald C. Figueiredo,
| |
Collapse
|
20
|
Nunes-Souza E, Silveira ME, Mendes MC, Nagashima S, de Paula CBV, da Silva GGVC, Barbosa GS, Martins JB, de Noronha L, Lenzi L, Barbosa JRS, Donin RDF, de Moura JF, Custódio G, Machado-Souza C, Lalli E, de Figueiredo BC. From adrenarche to aging of adrenal zona reticularis: precocious female adrenopause onset. Endocr Connect 2020; 9:1212-1220. [PMID: 33112833 PMCID: PMC7774755 DOI: 10.1530/ec-20-0416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/14/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Adaptive changes in DHEA and sulfated-DHEA (DHEAS) production from adrenal zona reticularis (ZR) have been observed in normal and pathological conditions. Here we used three different cohorts to assess timing differences in DHEAS blood level changes and characterize the relationship between early blood DHEAS reduction and cell number changes in women ZR. MATERIALS AND METHODS DHEAS plasma samples (n = 463) were analyzed in 166 healthy prepubertal girls before pubarche (<9 years) and 324 serum samples from 268 adult females (31.9-83.8 years) without conditions affecting steroidogenesis. Guided by DHEAS blood levels reduction rate, we selected the age range for ZR cell counting using DHEA/DHEAS and phosphatase and tensin homolog (PTEN), tumor suppressor and cell stress marker, immunostaining, and hematoxylin stained nuclei of 14 post-mortem adrenal glands. RESULTS We confirmed that overweight girls exhibited higher and earlier DHEAS levels and no difference was found compared with the average European and South American girls with a similar body mass index (BMI). Adrenopause onset threshold (AOT) defined as DHEAS blood levels <2040 nmol/L was identified in >35% of the females >40 years old and associated with significantly reduced ZR cell number (based on PTEN and hematoxylin signals). ZR cell loss may in part account for lower DHEA/DHEAS expression, but most cells remain alive with lower DHEA/DHEAS biosynthesis. CONCLUSION The timely relation between significant reduction of blood DHEAS levels and decreased ZR cell number at the beginning of the 40s suggests that adrenopause is an additional burden for a significant number of middle-aged women, and may become an emergent problem associated with further sex steroids reduction during the menopausal transition.
Collapse
Affiliation(s)
- Emanuelle Nunes-Souza
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
| | - Mônica Evelise Silveira
- Laboratório Central de Análises Clínicas, Hospital de Clínicas, Universidade Federal do Paraná, Centro, Curitiba, Paraná, Brazil
| | - Monalisa Castilho Mendes
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
| | - Seigo Nagashima
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, General Carneiro, Alto da Glória, Curitiba, Parana, Brazil
- Departamento de Medicina, PUC-PR, Prado Velho, Curitiba, Parana, Brazil
| | - Caroline Busatta Vaz de Paula
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, General Carneiro, Alto da Glória, Curitiba, Parana, Brazil
- Departamento de Medicina, PUC-PR, Prado Velho, Curitiba, Parana, Brazil
| | - Guilherme Guilherme Vieira Cavalcante da Silva
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, General Carneiro, Alto da Glória, Curitiba, Parana, Brazil
- Departamento de Medicina, PUC-PR, Prado Velho, Curitiba, Parana, Brazil
| | - Giovanna Silva Barbosa
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, General Carneiro, Alto da Glória, Curitiba, Parana, Brazil
- Departamento de Medicina, PUC-PR, Prado Velho, Curitiba, Parana, Brazil
| | - Julia Belgrowicz Martins
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
| | - Lúcia de Noronha
- Serviço de Anatomia Patológica, Hospital de Clínicas, Universidade Federal do Paraná, General Carneiro, Alto da Glória, Curitiba, Parana, Brazil
- Departamento de Medicina, PUC-PR, Prado Velho, Curitiba, Parana, Brazil
| | - Luana Lenzi
- Departamento de Análises Clínicas, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - José Renato Sales Barbosa
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
| | - Rayssa Danilow Fachin Donin
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
| | - Juliana Ferreira de Moura
- Pós Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica – UFPR, Curitiba, Brazil
| | - Gislaine Custódio
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
- Laboratório Central de Análises Clínicas, Hospital de Clínicas, Universidade Federal do Paraná, Centro, Curitiba, Paraná, Brazil
| | - Cleber Machado-Souza
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Sophia Antipolis, Valbonne, France
| | - Bonald Cavalcante de Figueiredo
- Pelé Pequeno Príncipe Research Institute, Água Verde, Curitiba, Parana, Brazil
- Faculdades Pequeno Príncipe, Rebouças, Curitiba, Parana, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at Universidade Federal do Paraná, Agostinho Leão Jr., Glória, Curitiba, Parana, Brazil
- Departamento de Saúde Coletiva, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
21
|
Jarzembowski JA. New Prognostic Indicators in Pediatric Adrenal Tumors: Neuroblastoma and Adrenal Cortical Tumors, Can We Predict When These Will Behave Badly? Surg Pathol Clin 2020; 13:625-641. [PMID: 33183724 DOI: 10.1016/j.path.2020.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pediatric adrenal tumors are unique entities with specific diagnostic, prognostic, and therapeutic challenges. The adrenal medulla gives rise to peripheral neuroblastic tumors (pNTs), pathologically defined by their architecture, stromal content, degree of differentiation, and mitotic-karyorrhectic index. Successful risk stratification of pNTs uses patient age, stage, tumor histology, and molecular/genetic aberrations. The adrenal cortex gives rise to adrenocortical tumors (ACTs), which present diagnostic and prognostic challenges. Histologic features that signify poor prognosis in adults can be meaningless in children, who have superior outcomes. The key clinical, pathologic, and molecular findings of pediatric ACTs have yet to be completely identified.
Collapse
Affiliation(s)
- Jason A Jarzembowski
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA; Pathology and Laboratory Medicine, Children's Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
22
|
Fu Y, Sun S, Bi J, Kong C. Construction of a risk signature for adrenocortical carcinoma using immune-related genes. Transl Androl Urol 2020; 9:1920-1930. [PMID: 33209656 PMCID: PMC7658150 DOI: 10.21037/tau-20-485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Adrenocortical carcinoma (ACC) is considered a rare tumor with a dismal prognosis. Expression of immune-related genes (IRGs) in ACC and correlations between IRGs and ACC prognosis were assessed using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Methods To preliminarily predict immune cell infiltration, an immune score was calculated using ESTIMATE. Differentially expressed IRGs were screened, and potential biological functions were investigated. We then performed univariate Cox regression to identify IRGs associated with survival, and the regulatory mechanisms of IRGs associated with survival were predicted. We built a risk signature through multivariate Cox regression to evaluate patient overall survival (OS). Results A high immune score predicted a good prognosis and an early clinical stage in ACC. We identified 30 IRGs associated with survival and then predicted associated regulatory mechanisms via protein-protein interaction (PPI) and transcription factor (TF) regulatory networks. The risk signature established by multivariate Cox regression correlated significantly with prognosis in ACC. Conclusions The vital roles of IRGs in ACC were assessed, and the risk signature obtained based on IRGs associated with survival independently predicted ACC prognosis.
Collapse
Affiliation(s)
- Yang Fu
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Shanshan Sun
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, China
| | - Jianbin Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Adrenocortical tumor (ACT) is a rare disease with an annual worldwide incidence of 0.3-0.38/million children below 15 years old, and Brazilian population presents the highest incidence because of germline mutation in the TP53. Pediatric ACT is associated with virilizing features and hypercortisolism in most cases. Malignancy is defined when local invasion or metastasis is found, and it is associated with a poor prognosis. However, the correct and early diagnosis and treatment may impact on overall and disease-free survival. RECENT FINDINGS A complete understanding of the disease and its singularities facilitates the assistance to the pediatric patient with ACT. The new insights about adrenal tumorigenesis have provided a better understanding of this disease. In this scenario, the era of molecular studies is leading to the refinement of the taxonomy, and it is offering the opportunity to discover new biomarkers and pathways of tumorigenesis, beyond the knowing β-catenin, Insulin-like growth factor-II/IGF-IR, and the p53/Rb signaling. SUMMARY The rarity of this disease makes it a real challenge. Here, we present a review focusing on clinical practice. A methodic approach aiming to clarify the diagnosis and a follow-up are suggested to guide physicians in the assistance of pediatrics patients, improving the prognosis.
Collapse
Affiliation(s)
- Vania B Brondani
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo
| | - Maria Candida B V Fragoso
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo
- Serviço de Endocrinologia da Clínica de Bases do Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| |
Collapse
|
24
|
Surakhy M, Wallace M, Bond E, Grochola LF, Perez H, Di Giovannantonio M, Zhang P, Malkin D, Carter H, Parise IZS, Zambetti G, Komechen H, Paraizo MM, Pagadala MS, Pinto EM, Lalli E, Figueiredo BC, Bond GL. A common polymorphism in the retinoic acid pathway modifies adrenocortical carcinoma age-dependent incidence. Br J Cancer 2020; 122:1231-1241. [PMID: 32147670 PMCID: PMC7156685 DOI: 10.1038/s41416-020-0764-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have enriched the fields of genomics and drug development. Adrenocortical carcinoma (ACC) is a rare cancer with a bimodal age distribution and inadequate treatment options. Paediatric ACC is frequently associated with TP53 mutations, with particularly high incidence in Southern Brazil due to the TP53 p.R337H (R337H) germline mutation. The heterogeneous risk among carriers suggests other genetic modifiers could exist. METHODS We analysed clinical, genotype and gene expression data derived from paediatric ACC, R337H carriers, and adult ACC patients. We restricted our analyses to single nucleotide polymorphisms (SNPs) previously identified in GWASs to associate with disease or human traits. RESULTS A SNP, rs971074, in the alcohol dehydrogenase 7 gene significantly and reproducibly associated with allelic differences in ACC age-of-onset in both cohorts. Patients homozygous for the minor allele were diagnosed up to 16 years earlier. This SNP resides in a gene involved in the retinoic acid (RA) pathway and patients with differing levels of RA pathway gene expression in their tumours associate with differential ACC progression. CONCLUSIONS These results identify a novel genetic component to ACC development that resides in the retinoic acid pathway, thereby informing strategies to develop management, preventive and therapeutic treatments for ACC.
Collapse
Affiliation(s)
- Mirvat Surakhy
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Marsha Wallace
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Elisabeth Bond
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Lukasz Filip Grochola
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Department of Surgery, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Husein Perez
- Faculty of Technology, Design and Environment, Oxford Brookes University, Oxford, UK
| | - Matteo Di Giovannantonio
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ping Zhang
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - David Malkin
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, USA
| | - Ivy Zortea S Parise
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Gerard Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heloisa Komechen
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Mariana M Paraizo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Meghana S Pagadala
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, USA
| | - Emilia M Pinto
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Université Côte D'Azur, Inserm, Valbonne, France
| | - Bonald C Figueiredo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil.
- Departamento de Saúde Coletiva, Universidade Federal do Paraná, Curitiba, PR, Brazil.
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Curitiba, PR, Brazil.
| | - Gareth L Bond
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Doghman-Bouguerra M, Finetti P, Durand N, Parise IZS, Sbiera S, Cantini G, Canu L, Hescot S, Figueiredo MMO, Komechen H, Sbiera I, Nesi G, Paci A, Al Ghuzlan A, Birnbaum D, Baudin E, Luconi M, Fassnacht M, Figueiredo BC, Bertucci F, Lalli E. Cancer-testis Antigen FATE1 Expression in Adrenocortical Tumors Is Associated with A Pervasive Autoimmune Response and Is A Marker of Malignancy in Adult, but Not Children, ACC. Cancers (Basel) 2020; 12:689. [PMID: 32183347 PMCID: PMC7140037 DOI: 10.3390/cancers12030689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
The SF-1 transcription factor target gene FATE1 encodes a cancer-testis antigen that has an important role in regulating apoptosis and response to chemotherapy in adrenocortical carcinoma (ACC) cells. Autoantibodies directed against FATE1 were previously detected in patients with hepatocellular carcinoma. In this study, we investigated the prevalence of circulating anti-FATE1 antibodies in pediatric and adult patients with adrenocortical tumors using three different methods (immunofluorescence, ELISA and Western blot). Our results show that a pervasive anti-FATE1 immune response is present in those patients. Furthermore, FATE1 expression is a robust prognostic indicator in adult patients with ACC and is associated with increased steroidogenic and decreased immune response gene expression. These data can open perspectives for novel strategies in ACC immunotherapy.
Collapse
Affiliation(s)
- Mabrouka Doghman-Bouguerra
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560 Valbonne, France; (M.D.-B.); (N.D.)
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
| | - Pascal Finetti
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, 232 Bd. Ste-Marguerite, 13009 Marseille, France; (P.F.); (D.B.); (F.B.)
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560 Valbonne, France; (M.D.-B.); (N.D.)
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
| | - Ivy Zortéa S. Parise
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
- Department, Pelé Pequeno Principe Research Institute, 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 2 Josef-Schneider-Straße, 97080 Würzburg, Germany; (S.S.); (I.S.); (M.F.)
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 6 Josef-Schneider-Straße, 97080 Würzburg, Germany
| | - Giulia Cantini
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 6 viale Pieraccini, 50139 Florence, Italy; (G.C.); (L.C.); (M.L.)
| | - Letizia Canu
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 6 viale Pieraccini, 50139 Florence, Italy; (G.C.); (L.C.); (M.L.)
| | - Ségolène Hescot
- Service de Médecine Nucléaire, Institut Curie, 35 rue Dailly, 92210 Saint Cloud, France;
| | - Mirna M. O. Figueiredo
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
- Department, Pelé Pequeno Principe Research Institute, 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil
| | - Heloisa Komechen
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
- Department, Pelé Pequeno Principe Research Institute, 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil
| | - Iuliu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 2 Josef-Schneider-Straße, 97080 Würzburg, Germany; (S.S.); (I.S.); (M.F.)
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 6 Josef-Schneider-Straße, 97080 Würzburg, Germany
| | - Gabriella Nesi
- Division of Pathological Anatomy, Department of Health Sciences, University of Florence, 6 viale Pieraccini, 50139 Florence, Italy;
| | - Angelo Paci
- Department of Neuro-Endocrine Tumors, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800 Villejuif, France; (A.P.); (A.A.G.); (E.B.)
| | - Abir Al Ghuzlan
- Department of Neuro-Endocrine Tumors, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800 Villejuif, France; (A.P.); (A.A.G.); (E.B.)
| | - Daniel Birnbaum
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, 232 Bd. Ste-Marguerite, 13009 Marseille, France; (P.F.); (D.B.); (F.B.)
| | - Eric Baudin
- Department of Neuro-Endocrine Tumors, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800 Villejuif, France; (A.P.); (A.A.G.); (E.B.)
| | - Michaela Luconi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 6 viale Pieraccini, 50139 Florence, Italy; (G.C.); (L.C.); (M.L.)
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 2 Josef-Schneider-Straße, 97080 Würzburg, Germany; (S.S.); (I.S.); (M.F.)
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 6 Josef-Schneider-Straße, 97080 Würzburg, Germany
| | - Bonald C. Figueiredo
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
- Department, Pelé Pequeno Principe Research Institute, 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil
| | - François Bertucci
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, 232 Bd. Ste-Marguerite, 13009 Marseille, France; (P.F.); (D.B.); (F.B.)
| | - Enzo Lalli
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France; 1532 Av. Silva Jardim, Curitiba PR 80250-200, Brazil; (I.Z.S.P.); (M.M.O.F.); (H.K.); (B.C.F.)
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des Lucioles - Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
26
|
Gao X, Yamazaki Y, Pecori A, Tezuka Y, Ono Y, Omata K, Morimoto R, Nakamura Y, Satoh F, Sasano H. Histopathological Analysis of Tumor Microenvironment and Angiogenesis in Pheochromocytoma. Front Endocrinol (Lausanne) 2020; 11:587779. [PMID: 33244312 PMCID: PMC7685215 DOI: 10.3389/fendo.2020.587779] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Pheochromocytomas (PHEOs) are relatively rare catecholamine-producing tumors derived from adrenal medulla. Tumor microenvironment (TME) including neoangiogenesis has been explored in many human neoplasms but not necessarily in PHEOs. Therefore, in this study, we examined tumor infiltrating lymphocytes (CD4 and CD8), tumor associated macrophages (CD68 and CD163), sustentacular cells (S100p), and angiogenic markers (CD31 and areas of intratumoral hemorrhage) in 39 cases of PHEOs in the quantitative fashion. We then compared the results with pheochromocytoma of the adrenal gland scaled score (PASS), grading system for pheochromocytoma and paraganglioma (GAPP) and the status of intra-tumoral catecholamine-synthesizing enzymes (TH, DDC, and PNMT) as well as their clinicopathological factors. Intratumoral CD8 (p = 0.0256), CD31 (p = 0.0400), and PNMT (p = 0.0498) status was significantly higher in PHEOs with PASS <4 than PASS ≧4. In addition, intratumoral CD8+ lymphocytes were also significantly more abundant in well-than moderately differentiated PHEO according to GAPP score (p = 0.0108) and inversely correlated with tumor size (p = 0.0257). Intratumoral CD68+ cells were significantly higher in PHEOs with regular or normal histological patterns than those not (p = 0.0370) and inversely correlated with tumor size (p = 0.0457). The status of CD163 was significantly positively correlated with that of CD8 positive cells (p = 0.0032). The proportion of intratumoral hemorrhage areas was significantly higher in PHEOs with PASS ≧4 (p = 0.0172). DDC immunoreactivity in tumor cells was significantly positively correlated with PASS score (p = 0.0356) and TH status was significantly higher in PHEOs harboring normal histological patterns (p = 0.0236) and cellular monotony (p = 0.0219) than those not. Results of our present study did demonstrate that abundant CD8+ and CD68+ cells could represent a histologically low-scored tumor. In particular, PHEOs with increased intratumoral hemorrhage should be considered rather malignant. In addition, abnormal catecholamine-producing status of tumor cells such as deficient PNMT and TH and increased DDC could also represent more aggressive PHEOs.
Collapse
Affiliation(s)
- Xin Gao
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alessio Pecori
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuta Tezuka
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yoshikiyo Ono
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Kei Omata
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Hironobu Sasano,
| |
Collapse
|