1
|
Michalski K, Schlötelburg W, Hartrampf PE, Kosmala A, Buck AK, Hahner S, Schirbel A. Radiopharmaceuticals for Treatment of Adrenocortical Carcinoma. Pharmaceuticals (Basel) 2023; 17:25. [PMID: 38256859 PMCID: PMC10820941 DOI: 10.3390/ph17010025] [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: 11/14/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Adrenocortical carcinoma (ACC) represents a rare tumor entity with limited treatment options and usually rapid tumor progression in case of metastatic disease. As further treatment options are needed and ACC metastases are sensitive to external beam radiation, novel theranostic approaches could complement established therapeutic concepts. Recent developments focus on targeting adrenal cortex-specific enzymes like the theranostic twin [123/131I]IMAZA that shows a good image quality and a promising therapeutic effect in selected patients. But other established molecular targets in nuclear medicine such as the C-X-C motif chemokine receptor 4 (CXCR4) could possibly enhance the therapeutic regimen as well in a subgroup of patients. The aims of this review are to give an overview of innovative radiopharmaceuticals for the treatment of ACC and to present the different molecular targets, as well as to show future perspectives for further developments since a radiopharmaceutical with a broad application range is still warranted.
Collapse
Affiliation(s)
- Kerstin Michalski
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| | - Wiebke Schlötelburg
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| | - Philipp E. Hartrampf
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| | - Aleksander Kosmala
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| | - Andreas K. Buck
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| | - Stefanie Hahner
- Division of Endocrinology and Diabetes, Department of Medicine I, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany;
| | - Andreas Schirbel
- Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany (A.K.B.); (A.S.)
| |
Collapse
|
2
|
Abstract
Adrenal cortical carcinoma (ACC) is a rare and aggressive malignancy that poses challenging issues regarding the diagnostic workup. Indeed, no presurgical technique or clinical parameters can reliably distinguish between adrenal cortical adenomas, which are more frequent and have a favorable outcome, and ACC, and the final diagnosis largely relies on histopathologic analysis of the surgical specimen. However, even the pathologic assessment of malignancy in an adrenal cortical lesion is not straightforward and requires a combined evaluation of multiple histopathologic features. Starting from the Weiss score, which was developed in 1984, several histopathologic scoring systems have been designed to tackle the difficulties of ACC diagnosis. Dealing with specific histopathologic variants (eg, Liss-Weiss-Bisceglia scoring system for oncocytic ACC) or patient characteristics (eg, Wieneke index in the pediatric setting), these scores remarkably improved the diagnostic workup of ACC and its subtypes. Nevertheless, cases with misleading features or discordant correlations between pathologic findings and clinical behavior still occur. Owing to multicentric collaborative studies integrating morphologic features with ancillary immunohistochemical markers and molecular analysis, ACC has eventually emerged as a multifaceted, heterogenous malignancy, and, while innovative and promising approaches are currently being tested, the future clinical management of patients with ACC will mainly rely on personalized medicine and target-therapy protocols. At the dawn of the new Fifth World Health Organization classification of endocrine tumors, this review will tackle ACC from the pathologist's perspective, thus focusing on the main available diagnostic, prognostic, and predictive tissue-tethered features and biomarkers and providing relevant clinical and molecular correlates.
Collapse
|
3
|
Ali Y, Gomez-Sanchez EP, Gomez-Sanchez CE. Mammalian Target of Rapamycin Inhibition Decreases Angiotensin II-Induced Steroidogenesis in HAC15 Human Adrenocortical Carcinoma Cells. Endocrinology 2022; 164:bqac185. [PMID: 36320101 PMCID: PMC9923797 DOI: 10.1210/endocr/bqac185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) inhibitors suppress adrenal cortical carcinoma cell proliferation and cortisol production; the relationship between mTOR and aldosterone production has not been examined. METHODS HAC15 cells were incubated with an mTOR activator and several inhibitors including AZD8055 (AZD) in the presence and absence of angiotensin II (AngII). The expression of rapamycin-sensitive adapter protein of mTOR (Raptor) and rapamycin-insensitive companion of mTOR (Rictor), adaptor proteins of mTOR complex 1 and 2, respectively, were studied in the HAC15 cells and deleted by CRISPR/gRNA. RESULTS The mTOR inhibitors decreased aldosterone induced by AngII. Inhibition of mTOR by AZD significantly suppressed AngII-induced aldosterone and cortisol formation in a dose-dependent manner, whereas the mTOR activator MHY had no effect. AZD did not alter forskolin-induced aldosterone production showing that it is specific to the AngII signaling pathway. AngII-mediated ERK and mTOR activation were suppressed by AZD, along with a concomitant dose-dependent reduction of AngII-induced steroidogenic enzymes including steroidogenic acute regulatory protein, 3β-hydroxysteroid dehydrogenase-type 2, CYP17A1, and aldosterone synthase protein. Furthermore, mTOR components ribosomal protein S6 kinase (P70S6K) and protein kinase B phosphorylation levels were decreased by AZD. As mTOR exerts its main effects by forming complexes with adaptor proteins Raptor and Rictor, the roles of these individual complexes were studied. We found an increase in the phosphorylation of Raptor and Rictor by AngII and that their CRISPR/gRNA-mediated knockdown significantly attenuated AngII-induced aldosterone and cortisol production. CONCLUSION mTOR signaling has a critical role in transducing the AngII signal initiating aldosterone and cortisol synthesis in HAC15 cells and that inhibition of mTOR could be a therapeutic option for conditions associated with excessive renin-angiotensin system-mediated steroid synthesis.
Collapse
Affiliation(s)
- Yusuf Ali
- G. V. (Sonny) Montgomery, VA Medical Center, Jackson, MS, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Celso E Gomez-Sanchez
- G. V. (Sonny) Montgomery, VA Medical Center, Jackson, MS, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
4
|
Cremaschi V, Abate A, Cosentini D, Grisanti S, Rossini E, Laganà M, Tamburello M, Turla A, Sigala S, Berruti A. Advances in adrenocortical carcinoma pharmacotherapy: what is the current state of the art? Expert Opin Pharmacother 2022; 23:1413-1424. [PMID: 35876101 DOI: 10.1080/14656566.2022.2106128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Surgery, followed or not by adjuvant mitotane, is the current mainstay of therapy for patients with early-stage adrenocortical carcinoma (ACC). Mitotane, either alone or in association with EDP (Etoposide-Doxorubicin-Cisplatin) combination chemotherapy, is the standard approach for patients with metastatic ACC. AREAS COVERED The activity of newer cytotoxic drugs, radioligands, targeted therapies and immunotherapy, both in preclinical and in clinical studies, will be reviewed in this paper. EXPERT OPINION ADIUVO trial revealed that the administration of adjuvant mitotane is not advantageous in patients with good prognosis. Future strategies are to intensify efforts in adjuvant setting in patients with high risk of relapse. In patients with advanced/metastatic disease, modern targeted therapies have shown significant cytotoxicity in preclinical studies, however, studies in ACC patients reported disappointing results so far. The absence of targeted agents specifically inhibiting the major molecular pathways of ACC growth is the main cause of the failure of these drugs. Since ACC is often antigenic but poorly immunogenic, the results of immunotherapy trials appeared inferior to those achieved in the management of patients with other malignancies. Radioligand therapy may also be a promising approach. Combination of chemotherapy plus immunotherapy could be interesting to be tested in the future.
Collapse
Affiliation(s)
- Valentina Cremaschi
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - Deborah Cosentini
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - Marta Laganà
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - Antonella Turla
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| |
Collapse
|
5
|
Steenaard RV, Ettaieb MHT, Kerkhofs TMA, Haak HR. How close are we to personalized mitotane dosing in the treatment of adrenocortical carcinoma? State of the art and future perspectives. Expert Opin Drug Metab Toxicol 2021; 17:677-683. [PMID: 33886381 DOI: 10.1080/17425255.2021.1921146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Mitotane is the only drug registered specifically for adrenocortical carcinoma. Finding the optimal dose for a patient is difficult due to large differences in bioavailability, toxicity and effect. We therefore look to improve personalized dosing of mitotane. AREAS COVERED We searched PubMed for studies related to mitotane dosing, pharmacokinetics, pharmacogenetics and combination therapy. Comparison of different dosing strategies have not resulted in an optimal advice. Several computerized pharmacokinetic models have been proposed to predict plasma levels. The current pharmacokinetic models do not explain the full variance in plasma levels. Pharmacogenetics have been proposed to find the unexplained variance. Studies on combination therapy have not yet led to a potential dose adjustment for mitotane. EXPERT OPINION Computerized pharmacokinetics models are promising tools to predict plasma levels, further validation is needed. Pharmacogenetics are introduced in these models, but more research is required before clinical application. We believe that in the near future, personalized mitotane dosage will be aided by a validated web-based pharmacokinetic model with good predictive ability based primarily on clinical characteristics, adjustable for actual plasma levels and dosage.
Collapse
Affiliation(s)
- Rebecca V Steenaard
- Department of Internal Medicine, Máxima MC, Veldhoven, Eindhoven, The Netherlands.,Maastricht University, CAPHRI School for Public Health and Primary Care, Ageing and Long-Term Care, Maastricht, The Netherlands
| | - Madeleine H T Ettaieb
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Thomas M A Kerkhofs
- Department of Internal Medicine, Division of Medical Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Harm R Haak
- Department of Internal Medicine, Máxima MC, Veldhoven, Eindhoven, The Netherlands.,Maastricht University, CAPHRI School for Public Health and Primary Care, Ageing and Long-Term Care, Maastricht, The Netherlands.,Department of Internal Medicine, Division of General Internal Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
6
|
Mizdrak M, Tičinović Kurir T, Božić J. The Role of Biomarkers in Adrenocortical Carcinoma: A Review of Current Evidence and Future Perspectives. Biomedicines 2021; 9:174. [PMID: 33578890 PMCID: PMC7916711 DOI: 10.3390/biomedicines9020174] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy arising from the adrenal cortex often with unexpected biological behavior. It can occur at any age, with two peaks of incidence: in the first and between fifth and seventh decades of life. Although ACC are mostly hormonally active, precursors and metabolites, rather than end products of steroidogenesis are produced by dedifferentiated and immature malignant cells. Distinguishing the etiology of adrenal mass, between benign adenomas, which are quite frequent in general population, and malignant carcinomas with dismal prognosis is often unfeasible. Even after pathohistological analysis, diagnosis of adrenocortical carcinomas is not always straightforward and represents a great challenge for experienced and multidisciplinary expert teams. No single imaging method, hormonal work-up or immunohistochemical labelling can definitively prove the diagnosis of ACC. Over several decades' great efforts have been made in finding novel reliable and available diagnostic and prognostic factors including steroid metabolome profiling or target gene identification. Despite these achievements, the 5-year mortality rate still accounts for approximately 75% to 90%, ACC is frequently diagnosed in advanced stages and therapeutic options are unfortunately limited. Therefore, imperative is to identify new biological markers that can predict patient prognosis and provide new therapeutic options.
Collapse
Affiliation(s)
- Maja Mizdrak
- Department of Nephrology and Hemodialysis, University Hospital of Split, 21000 Split, Croatia;
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| | - Tina Tičinović Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
- Department of Endocrinology, Diabetes and Metabolic Disorders, University Hospital of Split, 21000 Split, Croatia
| | - Joško Božić
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| |
Collapse
|
7
|
Ardolino L, Hansen A, Ackland S, Joshua A. Advanced Adrenocortical Carcinoma (ACC): a Review with Focus on Second-Line Therapies. HORMONES & CANCER 2020; 11:155-169. [PMID: 32303972 PMCID: PMC10355245 DOI: 10.1007/s12672-020-00385-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
Advanced adrenocortical cancer (ACC) is a rare, highly aggressive malignancy, which typically has a poor prognosis. In advanced ACC, the overall trend is toward a short PFS interval following first-line systemic therapy, highlighting a clear need for improved second-/third-line treatment strategies. We conducted a review of the literature and relevant scientific guidelines related to systemic therapy for advanced ACC. Public indexes including PubMed/MEDLINE were searched. Treatment selection in the second-line setting is based on small phase 2 trials, case reports, and pre-clinical evidence. The best data available for initial second-line therapy selection supports the use of gemcitabine and capecitabine (G + C) or streptozotocin (S), both with or without mitotane. G + C is becoming increasingly recommended based on phase 2 clinical trial data in patients of good PS, due to the inferred superior PFS and OS from non-comparative trials. Alternatively, streptozotocin was better tolerated than EDP + M in the FIRM-ACT study and remains an option when warranted. Beyond this, further treatment approaches should be tailored to individual patient characteristics, utilizing a mixture of systemic therapies, local therapies, and enrolment in clinical trials where available. Additionally, the role of molecular stratification, predictive biomarkers, and immune checkpoint inhibitors in specific individuals, such as Lynch syndrome, is evolving and may become increasingly utilized in clinical practice. Advanced ACC necessitates a multidisciplinary approach and is best managed in a specialist center. Although there is no one definitive second-line treatment strategy, there are some favorable approaches, which require further validation in larger clinical trials.
Collapse
Affiliation(s)
- Luke Ardolino
- The Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, NSW, Australia.
| | - Aaron Hansen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Stephen Ackland
- Calvary Mater Newcastle Hospital, University of Newcastle, Newcastle, NSW, Australia
| | - Anthony Joshua
- The Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, NSW, Australia
- St. Vincent's Clinical School, UNSW, Sydney, NSW, Australia
| |
Collapse
|
8
|
Grisanti S, Filice A, Basile V, Cosentini D, Rapa I, Albano D, Morandi A, Laganà M, Dalla Volta A, Bertagna F, Tiberio GMA, Volante M, Terzolo M, Versari A, Berruti A. Treatment With 90Y/177Lu-DOTATOC in Patients With Metastatic Adrenocortical Carcinoma Expressing Somatostatin Receptors. J Clin Endocrinol Metab 2020; 105:dgz091. [PMID: 31633759 DOI: 10.1210/clinem/dgz091] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/03/2019] [Accepted: 10/06/2019] [Indexed: 01/03/2023]
Abstract
CONTEXT We investigated the role of Gallium 68 dodecanetetraacetic acid Tyr3-octreotide (68Ga-DOTATOC) positron emission tomography/computed tomography (PET/CT) in detecting somatostatin receptors (SSTRs) in 19 patients with metastatic adrenocortical carcinoma (ACC) and explored the activity of yttrium-90/lutetium-177 (90Y/177Lu-DOTATOC) peptide receptor radionuclide therapy (PRRT). CASE DESCRIPTION AND METHODS 68Ga uptake in metastatic sites was scored in terms of intensity and anatomical uptake distribution of standard uptake value (SUV). Tissue expression of SSTR2A and SSTR5 was also evaluated by immunohistochemistry (IHC) on primary tumors. Eight (42%) patients displayed radiometabolic uptake of any-grade intensity with focal and limited distribution. Two (11%) patients displayed strong uptake in multiple lesions and were treated with PRRT. Both obtained an overall disease control lasting 4 and 12 months, respectively. CONCLUSIONS ACC can express SSTRs as detected by IHC and 68Ga-DOTATOC PET. SSTRs-based PRRT may represent a potential treatment opportunity for a minority of patients with advanced ACC. This treatment modality deserves further investigation.
Collapse
Affiliation(s)
- Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale - Istituto di Ricovero e Cura a Carattere Scientifico of Reggio Emilia, Reggio Emilia, Italy
| | - Vittoria Basile
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Hospital, University of Turin, Orbassano, Italy
| | - Deborah Cosentini
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| | - Ida Rapa
- Pathology Unit, Department of Oncology, S Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Domenico Albano
- Nuclear Medicine Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Morandi
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| | - Marta Laganà
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| | - Alberto Dalla Volta
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| | - Francesco Bertagna
- Nuclear Medicine Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Guido M A Tiberio
- Surgical Clinic, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Marco Volante
- Pathology Unit, Department of Oncology, S Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Massimo Terzolo
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Hospital, University of Turin, Orbassano, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale - Istituto di Ricovero e Cura a Carattere Scientifico of Reggio Emilia, Reggio Emilia, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia. ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
9
|
De Martino MC, Feelders RA, Pivonello C, Simeoli C, Papa F, Colao A, Pivonello R, Hofland LJ. The role of mTOR pathway as target for treatment in adrenocortical cancer. Endocr Connect 2019; 8:R144-R156. [PMID: 31398711 PMCID: PMC6733361 DOI: 10.1530/ec-19-0224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/08/2019] [Indexed: 12/23/2022]
Abstract
Adrenocortical carcinomas (ACCs) are rare tumors with scant treatment options for which new treatments are required. The mTOR pathway mediates the intracellular signals of several growth factors, including the insulin-like growth factors (IGFs), and therefore represents a potential attractive pathway for the treatment of several malignancies including ACCs. Several mTOR inhibitors, including sirolimus, temsirolimus and everolimus, have been clinically developed. This review summarizes the results of the studies evaluating the expression of the mTOR pathway components in ACCs, the effects of the mTOR inhibitors alone or in combination with other drugs in preclinical models of ACCs and the early experience with the use of these compounds in the clinical setting. The mTOR pathway seems a potential target for treatment of patients with ACC, but further investigation is still required to define the potential role of mTOR inhibitors alone or in combination with other drugs in the treatment of ACC patients.
Collapse
Affiliation(s)
- Maria Cristina De Martino
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Richard A Feelders
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Chiara Simeoli
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Fortuna Papa
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, Naples, Italy
| | - Leo J Hofland
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Wang X, Zhou N, Xiao Y, Zhu W, Bai C, Zhao L. Metastatic Adrenal Cortical Carcinoma Responding to Octreotide: A Case Report. Oncologist 2019; 24:e793-e797. [PMID: 31073023 DOI: 10.1634/theoncologist.2018-0855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 11/17/2022] Open
Abstract
Advanced adrenocortical carcinoma (ACC) is an aggressive disease with poor prognosis, and the current therapeutic options, such as mitotane or platinum-based chemotherapy regimens, often offer limited efficacy. Here, we present the first report, to the author's knowledge, of metastatic ACC with positive octreoscan scintigraphy that was successfully treated with octreotide long-acting release (LAR). A patient with metastatic ACC who showed poor tolerance to mitotane received octreotide LAR because of positive octreoscan scintigraphy. She obtained major partial response to the somatostatin analog. Interestingly, the expression of somatostatin receptor 2 from the previous local recurrence lesion was negative. The next-generation sequencing-based circulating tumor DNA analysis in the patient was performed and failed to identify any alterations. These findings suggest that octreotide LAR may be a good option for the treatment of metastatic ACC in selected patients.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Wenjia Zhu
- Departments of Nuclear Medicine and Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Lin Zhao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
11
|
McCabe MJ, Pinese M, Chan CL, Sheriff N, Thompson TJ, Grady J, Wong M, Gauthier MEA, Puttick C, Gayevskiy V, Hajdu E, Wong SQ, Barrett W, Earls P, Lukeis R, Cheng YY, Lin RCY, Thomas DM, Watkins DN, Dinger ME, McCormack AI, Cowley MJ. Genomic stratification and liquid biopsy in a rare adrenocortical carcinoma (ACC) case, with dual lung metastases. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003764. [PMID: 30936196 PMCID: PMC6549567 DOI: 10.1101/mcs.a003764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/11/2019] [Indexed: 12/22/2022] Open
Abstract
Adrenocortical carcinoma is a rare malignancy with a poor prognosis and few treatment options. Molecular characterization of this cancer remains limited. We present a case of an adrenocortical carcinoma (ACC) in a 37-yr-old female, with dual lung metastases identified 1 yr following commencement of adjuvant mitotane therapy. As standard therapeutic regimens are often unsuccessful in ACC, we undertook a comprehensive genomic study into this case to identify treatment options and monitor disease progress. We performed targeted and whole-genome sequencing of germline, primary tumor, and both metastatic tumors from this patient and monitored recurrence over 2 years using liquid biopsy for ctDNA and steroid hormone measurements. Sequencing revealed the primary and metastatic tumors were hyperhaploid, with extensive loss of heterozygosity but few structural rearrangements. Loss-of-function mutations were identified in MSH2, TP53, RB1, and PTEN, resulting in tumors with mismatch repair signatures and microsatellite instability. At the cellular level, tumors were populated by mitochondria-rich oncocytes. Longitudinal ctDNA mutation and hormone profiles were unable to detect micrometastatic disease, consistent with clinical indicators of disease remission. The molecular signatures in our ACC case suggested immunotherapy in the event of disease progression; however, the patient remains free of cancer. The extensive molecular analysis presented here could be applied to other rare and/or poorly stratified cancers to identify novel or repurpose existing therapeutic options, thereby broadly improving diagnoses, treatments, and prognoses.
Collapse
Affiliation(s)
- Mark J McCabe
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Mark Pinese
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Chia-Ling Chan
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Nisa Sheriff
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Department of Endocrinology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Tanya J Thompson
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - John Grady
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marie Wong
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marie-Emilie A Gauthier
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Clare Puttick
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Velimir Gayevskiy
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Elektra Hajdu
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Stephen Q Wong
- Molecular and Translational Genomics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Wade Barrett
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Peter Earls
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Robyn Lukeis
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Yuen Y Cheng
- Asbestos Diseases Research Institute, The University of Sydney, Sydney, New South Wales 2139, Australia
| | - Ruby C Y Lin
- Asbestos Diseases Research Institute, The University of Sydney, Sydney, New South Wales 2139, Australia.,Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, New South Wales 2145, Australia
| | - David M Thomas
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - D Neil Watkins
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Ann I McCormack
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia.,Department of Endocrinology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia.,Computational Biology Group, Children's Cancer Institute, Kensington, New South Wales 2031, Australia
| |
Collapse
|
12
|
Abstract
The diagnosis of low-grade adrenal cortical carcinoma (ACC) confined to the adrenal gland can be challenging. Although there are diagnostic and prognostic molecular tests for ACC, they remain largely unutilized. We examined the diagnostic and prognostic value of altered reticulin framework and the immunoprofile of biomarkers including IGF-2, proteins involved in cell proliferation and mitotic spindle regulation (Ki67, p53, BUB1B, HURP, NEK2), DNA damage repair (PBK, γ-H2AX), telomere regulation (DAX, ATRX), wnt-signaling pathway (beta-catenin) and PI3K signaling pathway (PTEN, phospho-mTOR) in a tissue microarray of 50 adenomas and 43 carcinomas that were characterized for angioinvasion as defined by strict criteria, Weiss score, and mitotic rate-based tumor grade. IGF-2 and proteins involved in cell proliferation and mitotic spindle regulation (Ki67, p53, BUB1B, HURP, NEK2), DNA damage proteins (PBK, γ-H2AX), regulators of telomeres (DAXX, ATRX), and beta-catenin revealed characteristic expression profiles enabling the distinction of carcinomas from adenomas. Not all biomarkers were informative in all carcinomas. IGF-2 was the most useful biomarker of malignancy irrespective of tumor grade and cytomorphologic features, as juxtanuclear Golgi-pattern IGF-2 reactivity optimized for high specificity was identified in up to 80% of carcinomas and in no adenomas. Loss rather than qualitative alterations of the reticulin framework yielded statistical difference between carcinoma and adenoma. Angioinvasion defined as tumor cells invading through a vessel wall and intravascular tumor cells admixed with thrombus proved to be the best prognostic parameter, predicting adverse outcome in the entire cohort as well as within low-grade ACCs. Low mitotic tumor grade, Weiss score, global loss of DAXX expression, and high phospho-mTOR expression correlated with disease-free survival, but Weiss score and biomarkers failed to predict adverse outcome in low-grade disease. Our results underscore the importance of careful morphologic assessment coupled with ancillary diagnostic and prognostic biomarkers of ACC.
Collapse
|
13
|
Crona J, Beuschlein F, Pacak K, Skogseid B. Advances in adrenal tumors 2018. Endocr Relat Cancer 2018; 25:R405-R420. [PMID: 29794126 PMCID: PMC5976083 DOI: 10.1530/erc-18-0138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022]
Abstract
This review aims to provide clinicians and researchers with a condensed update on the most important studies in the field during 2017. We present the academic output measured by active clinical trials and peer-reviewed published manuscripts. The most important and contributory manuscripts were summarized for each diagnostic entity, with a particular focus on manuscripts that describe translational research that have the potential to improve clinical care. Finally, we highlight the importance of collaborations in adrenal tumor research, which allowed for these recent advances and provide structures for future success in this scientific field.
Collapse
Affiliation(s)
- J Crona
- Department of Medical SciencesUppsala University, Uppsala, Sweden
| | - F Beuschlein
- Medizinische Klinik und Poliklinik IVKlinikum der Universität München, Munich, Germany
- Klinik für EndokrinologieDiabetologie und Klinische Ernährung, UniversitätsSpital Zürich, Zürich, Switzerland
| | - K Pacak
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - B Skogseid
- Department of Medical SciencesUppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Silveira E, Cavalcante IP, Kremer JL, de Mendonça POR, Lotfi CFP. The tyrosine kinase inhibitor nilotinib is more efficient than mitotane in decreasing cell viability in spheroids prepared from adrenocortical carcinoma cells. Cancer Cell Int 2018; 18:29. [PMID: 29507530 PMCID: PMC5831608 DOI: 10.1186/s12935-018-0527-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/19/2018] [Indexed: 02/01/2023] Open
Abstract
Background New drugs for adrenocortical carcinoma (ACC) are needed because most patients undergo rapid disease progression despite surgery and adjuvant therapy with mitotane. In this study, we aimed to investigate the in vitro effects of different chemotherapy drugs, alone or combined with mitotane, on the viability of adrenocortical carcinoma cells. Methods Everolimus, sunitinib, zoledronic acid, imatinib and nilotinib cytotoxicity, alone or combined with mitotane were tested on ACC H295R cells in monolayer or spheroid cultures using MTS assays and confocal microscopy. Moreover, the nilotinib effects were investigated in spheroids cultured from patient tumor-derived ACC-T36 cells. Results Morphological characterization of H295R cell spheroids using histochemistry was performed and showed that dense, homogenously sized, multicellular spheroids were obtained. We observed that sunitinib and nilotinib alone were equally effective in a monolayer preparation, whereas mitotane was the most effective even at a low dose. A combination of sunitinib and mitotane was the most effective treatment, with only 23.8% of cells in the monolayer remaining viable. Spheroid preparations showed resistance to different drugs, although the poor effect produced by mitotane alone was surprising, with a cell viability of 84.6% in comparison with 13.1% in monolayer cells. The most ineffective drugs in spheroid preparations were everolimus, zoledronic acid and imatinib. In both cell types, nilotinib, either alone or in combination with mitotane induced more significant cell viability inhibition in monolayer and spheroid preparations. In addition, the mechanism of nilotinib activity involves the ERK1/2 pathway. Conclusion Taken together, our data identified nilotinib as a cytotoxic drug that combined with ERK inhibitors deserves to be tested as a novel therapy for adrenocortical carcinoma.
Collapse
Affiliation(s)
- Elaine Silveira
- Department of Anatomy, Institute of Biomedical Science, University of São Paulo, São Paulo, SP Brazil
| | - Isadora Pontes Cavalcante
- Department of Anatomy, Institute of Biomedical Science, University of São Paulo, São Paulo, SP Brazil
| | - Jean Lucas Kremer
- Department of Anatomy, Institute of Biomedical Science, University of São Paulo, São Paulo, SP Brazil
| | | | | |
Collapse
|
15
|
Kieler M, Müllauer L, Koperek O, Bianconi D, Unseld M, Raderer M, Prager GW. Analysis of 10 Adrenocortical Carcinoma Patients in the Cohort of the Precision Medicine Platform MONDTI. Oncology 2018; 94:306-310. [PMID: 29444511 DOI: 10.1159/000486678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/22/2017] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Adrenocortical carcinoma (ACC) is a rare disease with a dismal prognosis. We aimed to evaluate if a personalized medicine approach may be useful for matching patients with ACC to targeted therapies. METHODS This is an analysis of 10 molecularly profiled ACCs that were progressing under standard of care treatment. The profile consisted of a 50-gene next-generation sequencing panel, immunohistochemistry (IHC), and fluorescence in situ hybridization for several proteins or chromosomal aberrations. RESULTS In 6 (60%) tumor samples, no somatic mutation was detected, while in 3 (30%) tumors 1 mutation was detected and in 1 (10%) tumor 2 mutations were detected. These mutations were CTNNB1 (2 samples), TP53 (1 sample), RB1 (1 sample) and APC (1 sample). Expression of phospho-mTOR and of EGFR was commonly detected by IHC (87.5 and 62.5%). In 4 (50%) samples, IHC revealed a weak expression of progesterone receptor. Less frequent alterations were expression of PDGFR-α, c-KIT, and estrogen receptor, each in 1 case. CONCLUSIONS Based on the molecular profile, no recommendation for targeted therapy was made by the multi-disciplinary team. Currently, ACC might not be suitable for a precision medicine approach according to our tests.
Collapse
Affiliation(s)
- Markus Kieler
- Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Leonhard Müllauer
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Oskar Koperek
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Daniela Bianconi
- Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Matthias Unseld
- Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Markus Raderer
- Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Gerald W Prager
- Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| |
Collapse
|