Next Article in Journal
Nuclear Cross-Section of Proton-Induced Reactions on Enriched 48Ti Targets for the Production of Theranostic 47Sc Radionuclide, 46cSc, 44mSc, 44gSc, 43Sc, and 48V
Next Article in Special Issue
Alpha-Emitting Radionuclides: Current Status and Future Perspectives
Previous Article in Journal
Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
Previous Article in Special Issue
Combining Cisplatin with Different Radiation Qualities—Interpretation of Cytotoxic Effects In Vitro by Isobolographic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiopharmaceuticals for Treatment of Adrenocortical Carcinoma

by
Kerstin Michalski
1,*,
Wiebke Schlötelburg
1,
Philipp E. Hartrampf
1,
Aleksander Kosmala
1,
Andreas K. Buck
1,
Stefanie Hahner
2 and
Andreas Schirbel
1
1
Department of Nuclear Medicine, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany
2
Division of Endocrinology and Diabetes, Department of Medicine I, Würzburg University Hospital, University of Würzburg, Oberdürrbacher Straße 6, D-97080 Würzburg, Germany
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(1), 25; https://doi.org/10.3390/ph17010025
Submission received: 14 November 2023 / Revised: 20 December 2023 / Accepted: 21 December 2023 / Published: 23 December 2023
(This article belongs to the Special Issue Therapeutic Radionuclides in Nuclear Medicine)

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.

1. Introduction

Adrenocortical carcinoma (ACC) is a rare tumor entity with an estimated incidence of about 0.5–2 new cases per million people per year [1,2]. ACC occurs at any age and shows a peak incidence between 40 and 60 years, whereby women are more often affected (55–60%) [3]. The tumor arises from the cortex of the adrenal gland and 50–60% of patients with ACC have clinical hormone excess. Treatment options are limited and complete resection is the only means of cure. Still, retrospective studies reported that 40–70% of ACCs eventually recur even after complete resection [4,5,6,7]. In general, the prognosis is heterogeneous and the median overall survival of all ACC patients is about 3–4 years. For tumors confined to the adrenal gland five-year survival rates are between 60–80%, for locally advanced disease 35–50%, and much lower in case of metastases with reported survival rates ranging from 0 to 28% [8,9,10,11,12,13,14].
Due to the rareness of the disease and the limited resources dedicated to the implementation of new therapeutic options, there is little progress in the medical therapy of ACC [15]. International guidelines recommend to use of adjuvant mitotane in most patients [3,16]. The results of a large phase 3 trial led to a combination treatment of mitotane, etoposide, doxorubicin, and cisplatin as a first-line therapy [11]. Unfortunately, the combination of these chemotherapeutics only led to an objective response rate of 23% with a progression-free survival of only 5.1 months despite severe toxicity. Hence, further therapeutic options for second- and third-line treatment are warranted. ACC used to be considered resistant to radiation [17,18]. However, recent data show a benefit in regards to local tumor control, the palliative treatment of symptomatic cerebral or osseous metastases and in case of vena cava obstruction as well as a reduction of local recurrence after primary resection [19,20,21,22,23,24,25]. In this sense, endoradiotherapy is a possible therapeutic option in patients with metastasized ACC after first-line treatment. The concept of endoradiotherapy is based upon theranostic radiopharmaceuticals that can be used for diagnostic and therapeutic purposes, depending on the labeled radionuclide. It is possible to use either the same molecule or a very similar compound. These molecules are radiolabeled with gamma and positron emitters for imaging purposes or beta minus emitters and (rarer) alpha emitters for endoradiotherapy. Some radionuclides, such as iodine-131 and lutetium-177 are beta and gamma emitters and can be used for both imaging and therapy, whereas the gamma emitter iodine-123 can be used only for diagnostics [26]. Other radionuclides for imaging are fluorine-18 or gallium-68 (both positron emitters) and Yttrium-90 (beta minus emitter) for therapy. The use of an image-based patient selection allows for a personalized medicine approach with a possible higher therapeutic efficacy. Furthermore, reduced side effects and high tumor doses can be administered because of the precise radiation deposition and the short tissue penetration of only a few millimeters of beta minus emitters [27].
The present review aims to give an overview of theranostic radiopharmaceuticals for the treatment of ACC, to present various molecular targets and to show future perspectives.

2. Molecular Imaging and Theranostic Approaches in ACC

For molecular imaging of ACC, positron emission tomography (PET)/computed tomography (CT) with [18F]fluorodeoxyglucose (FDG) can be used [28] but is not considered standard of care [3], in contrast to CT or magnetic resonance imaging. Nevertheless, FDG PET/CT is useful for prognostic evaluation as a higher uptake is associated with a shorter survival [29,30]. However, FDG does not provide a theranostic approach. For a detailed description of molecular imaging approaches in ACC, please refer to a recent review of adrenal imaging [31].
Peptide receptor radionuclide therapy targeting the somatostatin receptor (SSTR) using, i.e., [177Lu]Lu-DOTA-0-Tyr3-Octreotate (DOTATATE) is established in the treatment of well-differentiated neuroendocrine midgut tumors [32] and other neuroendocrine tumors [33]. A recent ex vivo study described a heterogeneous SSTR expression in some ACC tissue samples [34]. However, to date, only one study exists that reports the results of a case series of 19 patients with 2 patients receiving either [90Y]Y- or [177Lu]Lu-DOTATOC (DOTA(0)-Phe(1)-Tyr(3))octreotid), which resulted in disease control of 4 and 12 months, respectively [35].
In analogy, endoradiotherapy targeting the prostate-specific membrane antigen (PSMA) is not just a treatment option for metastasized castration-resistant prostate cancer using, i.e., [177Lu]Lu-vipivotide tetraxetan (PSMA-617) [36], but also for other tumor entities. In an ex vivo analysis, PSMA was significantly overexpressed in ACC tissue samples compared to normal adrenal glands and adrenocortical adenomas [37]. To our knowledge, there is no report providing data on PSMA radioligand therapy in ACC. Only one case report describes a patient with ACC having a PSMA expression in tumor sites equal to physiological liver background on [68Ga]Ga-PSMA-11 PET/CT, which was not considered sufficient for PSMA-directed radioligand therapy [38].
C-X-C motif chemokine receptor 4 (CXCR4) is a G-protein coupled receptor that can be found in many hematological malignancies as well as solid tumors and constitutes a possible theranostic target [39]. CXCR4 expression can be found in ACC samples as well [40]. A strong membranous expression of CXCR4 in ACC specimens was found in half of the cases (94 of 187 specimens) in an ex vivo study. Interestingly, immunohistochemical staining of CXCR4 was higher in samples derived from metastases than from primary tumors [41]. A high in vivo CXCR4 expression on CXCR4-directed PET/CT was found in 30 patients with ACC [42]. A possible theranostic application was found by Bluemel et al., who rated 17 (57%) of 30 patients as suitable and 4 patients (13%) as potentially suitable for CXCR4-directed treatment [43]. Of note, CXCR4-directed therapy using, i.e., [177Lu]Lu-/[90Y]Y-anditixafortide (PentixaTher) leads to bone marrow ablation and can only be applied in case of available hematopoietic stem cells which are usually harvested during previous chemotherapeutic protocols [44].
The enzymes CYP11B1 (11β-hydroxylase) and CYP11B2 (aldosterone synthase) are part of the cortisol and aldosterone synthesis in the adrenal gland and can be blocked by imidazole drugs such as etomidate or ketoconazole [45]. As these enzymes are highly specific for the adrenal gland, they are potential targets for molecular imaging [46]. Bergström et al. developed the PET imaging agent [11C]etomidate and its methyl ester [11C]metomidate ([11C]MTO) and showed their potential to specifically visualize the normal adrenal cortex in an animal study [47]. This approach was transferred to a clinical setting and the authors could demonstrate that [11C]MTO PET can distinguish between lesions of adrenocortical and nonadrenocortical origin in a cohort of 15 patients [48], and in another cohort of 173 patients [49]. The latter study included 13 patients with ACC which showed a relatively high tracer uptake.
In order to develop a possible theranostic radiopharmaceutical, the compound [123I]iodometomidate ([123I]IMTO) that inhibits CYP11B1/2 was developed. High imaging quality was shown in animal studies [50,51,52] and a high and specific tracer uptake of the radiopharmaceutical was found for adrenocortical tissue [51]. These promising results could be transferred into clinical application: [123I]IMTO planar whole-body scans and single photon emission computed tomography (SPECT)/CT images showed high sensitivity and specificity for the differentiation of adrenocortical tumors from lesions of non-adrenocortical origin in case of a lesion size of 2 cm or more [53]. The theranostic counterpart of [123I]IMTO is [131I]IMTO, which can be used in patients with advanced ACC. Disease control was achieved in 6 of 11 patients with ACC treated with [131I]IMTO with a median progression-free survival of 14 months (range 5–33 months) in responders. Of these, 5 patients showed a stable disease on follow-up CT scans, and a partial response was found in one patient [54]. As IMTO shows a rapid metabolic inactivation, the metabolically more stable derivative (R)-1-[1-(4-iodophenyl)ethyl]-1H-imidazole-5-carboxylic acid azetidinyl amide (IMAZA) was developed by replacing the methyl ester in IMTO by a carboxylic amide. IMAZA outperformed IMTO in regards to pharmacokinetic and imaging properties in mice and in a dual tracer approach in three patients [55]. Hahner et al. screened 69 patients with advanced ACC refractory to standard treatments using [123I]IMAZA SPECT/CT and identified 13 patients with intense uptake in all tumor lesions [56]. These patients were treated with a median of 25.7 GBq [131I]IMAZA (range 18.1–30.7 GBq). Response to therapy was assessed according to Response Evaluation Criteria in Solid Tumors (RECIST version 1.1) [57]. Two patients experienced a decrease in RECIST target lesions of up to 26%. A median progression-free survival of 14.3 months (range 8.3–21.9) was noted for five patients with stable disease. Median overall survival in all 13 patients was 14.1 months (4.0–56.5). The treatment was well tolerated by the patients, and no severe toxicities (CTCAE grade ≥ 3) were noted. Figure 1 shows a patient who underwent [131I]IMAZA therapy. Figure 2 summarizes the different theranostic targets in ACC and Figure 3 shows the corresponding radiopharmaceuticals.

3. Radiosynthesis of [131I]IMAZA

The radiosynthesis and quality control of [123/131I]IMAZA for scintigraphy, dosimetry and therapy has already been published [55]. Here, destannylation reactions were used for labeling. Since this method yields the labeled products under very mild reaction conditions and with very high radiochemical yields, this method is frequently used and should be easily established in radiochemical laboratories that have experience with radioiodination. However, this does not apply to radioiodinations with > 30 GBq I-131, which are challenging in terms of radiation protection due to the high volatility of radioiodine in combination with the extremely high activity levels and the relatively high gamma energy of 364 keV. Therefore, labeling of [131I]IMAZA for endoradiotherapy had to be performed by an automated synthesis module (custom-made by Scintomics GmbH, Fürstenfeldbruck, Germany) inside a well-ventilated lead cell (see Figure 4).
To the delivery vial in which the [131I]iodide is dissolved in 1 mL 0.01 N NaOH (IBSSO; GE Healthcare, Braunschweig, Germany) were consecutively injected 5 mg trimethylstannylazetidinylamide in 1 mL ethanol, 120 µL 2 N hydrochloric acid and 2.25 mg chloramine T trihydrate in 150 µL water. The reaction solution was allowed to stand for three minutes. Thereafter, the reaction was quenched by adding 135 µL 2 N HCl and a solution of 4.50 mg Na2S2O5 in 150 µL water and the mixture was injected directly into the injection valve of the semi-preparative high-performance liquid chromatography system (HPLC) equipped with a RP-18 HPLC column (250 × 8 mm). An ethanol/phosphate buffer (40/60 v/v) mixture served as the HPLC solvent with a flow of 2.0 mL/min. Using typical starting activities of 34 GBq [131I]iodide, reproducibly > 25 GBq [131I]IMAZA were obtained, which were administered to the patients after successful quality control.
For each radiosynthesis, the exhaust air from the lead box was passed through activated carbon filters and checked for possible contamination. The personnel involved were monitored by means of personal dosimeters, finger ring dosimeters and a thyroid monitor. In all cases, only very low levels of contamination were detectable, so that the high-dose endoradiotherapies with [131I]IMAZA could be carried out safely. Regarding the radiosynthesis of the commercially available products [177Lu]Lu-DOTATATE, [177Lu]Lu-/[90Y]Y-PentixaTher and [177Lu]Lu-PSMA-617, please refer to the respective publications [58,59,60,61].

4. Future Perspectives

The investigations of patients with metastatic ACC with [123I]IMAZA showed an uptake in all known lesions (metastases and/or primary tumor) in only about 40% of the patients. This is likely due to dedifferentiation of the tumor cells resulting in low or no expression of the target enzymes CYP11B1 and CYP11B2. Therefore, only a minority of patients with high tracer uptake are candidates for subsequent endoradiotherapy with the analog [131I]IMAZA. Currently, alternative enzymatic and non-enzymatic targets with broader expression in ACC tissue are under investigation.

5. Summary

Adrenocortical carcinoma is a rare tumor entity and further therapeutic options in metastatic disease are desperately warranted. Several possible theranostic approaches exist, of which radiopharmaceuticals targeting specific enzymes of the adrenal cortex are currently the most promising and are the only theranostic radiopharmaceuticals ever used in patients to date. The theranostic twin [123/131I]IMAZA has shown good image quality and a good therapeutic effect in selected patients with advanced ACC, but cannot be used in all patients with ACC. Therefore, future developments are needed in order to provide a radiopharmaceutical with broader applications.

Funding

This work was supported by the Bavarian Cancer Research Center (personal grant to K.M.) and by the Interdisciplinary Center of Clinical Research (IZKF), University Hospital of Wuerzburg (grant Z-2/91 to W.S.).

Data Availability Statement

Data sharing is not applicable.

Conflicts of Interest

K.M. has received speaker honoraria from Novartis. All other authors declare no conflicts of interest.

References

  1. Kerkhofs, T.M.A.; Verhoeven, R.H.A.; Van der Zwan, J.M.; Dieleman, J.; Kerstens, M.N.; Links, T.P.; Van de Poll-Franse, L.V.; Haak, H.R. Adrenocortical carcinoma: A population-based study on incidence and survival in the Netherlands since 1993. Eur. J. Cancer 2013, 49, 2579–2586. [Google Scholar] [CrossRef] [PubMed]
  2. Kebebew, E.; Reiff, E.; Duh, Q.Y.; Clark, O.H.; McMillan, A. Extent of disease at presentation and outcome for adrenocortical carcinoma: Have we made progress? World J. Surg. 2006, 30, 872–878. [Google Scholar] [CrossRef] [PubMed]
  3. Fassnacht, M.; Dekkers, O.M.; Else, T.; Baudin, E.; Berruti, A.; de Krijger, R.; Haak, H.R.; Mihai, R.; Assie, G.; Terzolo, M. European Society of Endocrinology Clinical Practice Guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Network for the Study of Adrenal Tumors. Eur. J. Endocrinol. 2018, 179, G1–G46. [Google Scholar] [CrossRef] [PubMed]
  4. Beuschlein, F.; Weigel, J.; Saeger, W.; Kroiss, M.; Wild, V.; Daffara, F.; Libe, R.; Ardito, A.; Al Ghuzlan, A.; Quinkler, M.; et al. Major prognostic role of Ki67 in localized adrenocortical carcinoma after complete resection. J. Clin. Endocrinol. Metab. 2015, 100, 841–849. [Google Scholar] [CrossRef] [PubMed]
  5. Glenn, J.A.; Else, T.; Hughes, D.T.; Cohen, M.S.; Jolly, S.; Giordano, T.J.; Worden, F.P.; Gauger, P.G.; Hammer, G.D.; Miller, B.S. Longitudinal patterns of recurrence in patients with adrenocortical carcinoma. Surgery 2019, 165, 186–195. [Google Scholar] [CrossRef] [PubMed]
  6. Liang, J.; Liu, Z.; Zhou, L.; Tang, Y.; Zhou, C.; Wu, K.; Zhang, F.; Zhang, F.; Wei, X.; Lu, Y.; et al. The clinical utility of ‘GRAS’ parameters in stage I-III adrenocortical carcinomas: Long-term data from a high-volume institution. Endocrine 2020, 67, 449–456. [Google Scholar] [CrossRef] [PubMed]
  7. Elhassan, Y.S.; Altieri, B.; Berhane, S.; Cosentini, D.; Calabrese, A.; Haissaguerre, M.; Kastelan, D.; Fragoso, M.; Bertherat, J.; Al Ghuzlan, A.; et al. S-GRAS score for prognostic classification of adrenocortical carcinoma: An international, multicenter ENSAT study. Eur. J. Endocrinol. 2021, 186, 25–36. [Google Scholar] [CrossRef] [PubMed]
  8. Fassnacht, M.; Libe, R.; Kroiss, M.; Allolio, B. Adrenocortical carcinoma: A clinician’s update. Nat. Rev. Endocrinol. 2011, 7, 323–335. [Google Scholar] [CrossRef]
  9. Fassnacht, M.; Johanssen, S.; Fenske, W.; Weismann, D.; Agha, A.; Beuschlein, F.; Fuhrer, D.; Jurowich, C.; Quinkler, M.; Petersenn, S.; et al. Improved Survival in Patients with Stage II Adrenocortical Carcinoma Followed Up Prospectively by Specialized Centers. J. Clin. Endocr. Metab. 2010, 95, 4925–4932. [Google Scholar] [CrossRef]
  10. Fassnacht, M.; Johanssen, S.; Quinkler, M.; Bucsky, P.; Willenberg, H.S.; Beuschlein, F.; Terzolo, M.; Mueller, H.H.; Hahner, S.; Allolio, B.; et al. Limited Prognostic Value of the 2004 International Union Against Cancer Staging Classification for Adrenocortical Carcinomas. Cancer 2009, 115, 243–250. [Google Scholar] [CrossRef]
  11. Fassnacht, M.; Terzolo, M.; Allolio, B.; Baudin, E.; Haak, H.; Berruti, A.; Welin, S.; Schade-Brittinger, C.; Lacroix, A.; Jarzab, B.; et al. Combination chemotherapy in advanced adrenocortical carcinoma. N. Engl. J. Med. 2012, 366, 2189–2197. [Google Scholar] [CrossRef] [PubMed]
  12. Sturgeon, C.; Shen, W.T.; Clark, O.H.; Duh, Q.Y.; Kebebew, E. Risk assessment in 457 adrenal cortical carcinomas: How much does tumor size predict the likelihood of malignancy? J. Am. Coll. Surg. 2006, 202, 423–430. [Google Scholar] [CrossRef] [PubMed]
  13. Bilimoria, K.Y.; Shen, W.T.; Elaraj, D.; Bentrem, D.J.; Winchester, D.J.; Kebebew, E.; Sturgeon, C. Adrenocortical carcinoma in the United States: Treatment utilization and prognostic factors. Cancer 2008, 113, 3130–3136. [Google Scholar] [CrossRef] [PubMed]
  14. Kerkhofs, T.M.; Ettaieb, M.H.; Hermsen, I.G.; Haak, H.R. Developing treatment for adrenocortical carcinoma. Endocr. Relat. Cancer 2015, 22, R325–R338. [Google Scholar] [CrossRef] [PubMed]
  15. Terzolo, M.; Fassnacht, M. Our experience with the management of patients with non-metastatic adrenocortical carcinoma. Eur. J. Endocrinol. 2022, 187, R27–R40. [Google Scholar] [CrossRef] [PubMed]
  16. Fassnacht, M.; Assie, G.; Baudin, E.; Eisenhofer, G.; de la Fouchardiere, C.; Haak, H.R.; de Krijger, R.; Porpiglia, F.; Terzolo, M.; Berruti, A.; et al. Adrenocortical carcinomas and malignant phaeochromocytomas: ESMO-EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020, 31, 1476–1490. [Google Scholar] [CrossRef] [PubMed]
  17. Hutter, A.M., Jr.; Kayhoe, D.E. Adrenal cortical carcinoma. Clinical features of 138 patients. Am. J. Med. 1966, 41, 572–580. [Google Scholar] [CrossRef] [PubMed]
  18. Hajjar, R.A.; Hickey, R.C.; Samaan, N.A. Adrenal cortical carcinoma. A study of 32 patients. Cancer 1975, 35, 549–554. [Google Scholar] [CrossRef]
  19. Polat, B.; Fassnacht, M.; Pfreundner, L.; Guckenberger, M.; Bratengeier, K.; Johanssen, S.; Kenn, W.; Hahner, S.; Allolio, B.; Flentje, M. Radiotherapy in adrenocortical carcinoma. Cancer 2009, 115, 2816–2823. [Google Scholar] [CrossRef]
  20. Hermsen, I.G.; Groenen, Y.E.; Dercksen, M.W.; Theuws, J.; Haak, H.R. Response to radiation therapy in adrenocortical carcinoma. J. Endocrinol. Investig. 2010, 33, 712–714. [Google Scholar] [CrossRef]
  21. Ho, J.; Turkbey, B.; Edgerly, M.; Alimchandani, M.; Quezado, M.; Camphausen, K.; Fojo, T.; Kaushal, A. Role of radiotherapy in adrenocortical carcinoma. Cancer J. 2013, 19, 288–294. [Google Scholar] [CrossRef] [PubMed]
  22. Fassnacht, M.; Hahner, S.; Polat, B.; Koschker, A.C.; Kenn, W.; Flentje, M.; Allolio, B. Efficacy of adjuvant radiotherapy of the tumor bed on local recurrence of adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2006, 91, 4501–4504. [Google Scholar] [CrossRef] [PubMed]
  23. Sabolch, A.; Feng, M.; Griffith, K.; Hammer, G.; Doherty, G.; Ben-Josef, E. Adjuvant and definitive radiotherapy for adrenocortical carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2011, 80, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, J.; Zheng, Z.; Shen, J.; Lian, X.; Miao, Z.; Shen, J.; Zhang, F. Efficacy of adjuvant radiotherapy for treatment of adrenocortical carcinoma: A retrospective study and an updated meta-analysis. Radiat. Oncol. 2020, 15, 118. [Google Scholar] [CrossRef] [PubMed]
  25. Gharzai, L.A.; Green, M.D.; Griffith, K.A.; Else, T.; Mayo, C.S.; Hesseltine, E.; Spratt, D.E.; Ben-Josef, E.; Sabolch, A.; Miller, B.S.; et al. Adjuvant Radiation Improves Recurrence-Free Survival and Overall Survival in Adrenocortical Carcinoma. J. Clin. Endocrinol. Metab. 2019, 104, 3743–3750. [Google Scholar] [CrossRef] [PubMed]
  26. Yordanova, A.; Eppard, E.; Kurpig, S.; Bundschuh, R.A.; Schonberger, S.; Gonzalez-Carmona, M.; Feldmann, G.; Ahmadzadehfar, H.; Essler, M. Theranostics in nuclear medicine practice. Onco Targets Ther. 2017, 10, 4821–4828. [Google Scholar] [CrossRef] [PubMed]
  27. Kramer-Marek, G.; Capala, J. The role of nuclear medicine in modern therapy of cancer. Tumour Biol. 2012, 33, 629–640. [Google Scholar] [CrossRef]
  28. Han, S.J.; Kim, T.S.; Jeon, S.W.; Jeong, S.J.; Yun, M.; Rhee, Y.; Kang, E.S.; Cha, B.S.; Lee, E.J.; Lee, H.C.; et al. Analysis of adrenal masses by F-18-FDG positron emission tomography scanning. Int. J. Clin. Pr. 2007, 61, 802–809. [Google Scholar] [CrossRef]
  29. Leboulleux, S.; Dromain, C.; Bonniaud, G.; Auperin, A.; Caillou, B.; Lumbroso, J.; Sigal, R.; Baudin, E.; Schlumberger, M. Diagnostic and prognostic value of 18-fluorodeoxyglucose positron emission tomography in adrenocortical carcinoma: A prospective comparison with computed tomography. J. Clin. Endocr. Metab. 2006, 91, 920–925. [Google Scholar] [CrossRef]
  30. Wrenn, S.M.; Moore, A.L.; Shah, H.J.; Barletta, J.A.; Vaidya, A.; Kilbridge, K.L.; Doherty, G.M.; Jacene, H.A.; Nehs, M.A. Higher SUVmax on FDG-PET is associated with shorter survival in adrenocortical carcinoma. Am. J. Surg. 2023, 225, 309–314. [Google Scholar] [CrossRef]
  31. Werner, R.A.; Hartrampf, P.E.; Schirbel, A.; Hahner, S. Adrenal functional imaging—Which marker for which indication? Curr. Opin. Urol. 2022, 32, 585–593. [Google Scholar] [CrossRef] [PubMed]
  32. Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef] [PubMed]
  33. Camus, B.; Cottereau, A.S.; Palmieri, L.J.; Dermine, S.; Tenenbaum, F.; Brezault, C.; Coriat, R. Indications of Peptide Receptor Radionuclide Therapy (PRRT) in Gastroenteropancreatic and Pulmonary Neuroendocrine Tumors: An Updated Review. J. Clin. Med. 2021, 10, 1267. [Google Scholar] [CrossRef] [PubMed]
  34. Germano, A.; Rapa, I.; Duregon, E.; Votta, A.; Giorcelli, J.; Buttigliero, C.; Scagliotti, G.V.; Volante, M.; Terzolo, M.; Papotti, M. Tissue Expression and Pharmacological In Vitro Analyses of mTOR and SSTR Pathways in Adrenocortical Carcinoma. Endocr. Pathol. 2017, 28, 95–102. [Google Scholar] [CrossRef] [PubMed]
  35. Grisanti, S.; Filice, A.; Basile, V.; Cosentini, D.; Rapa, I.; Albano, D.; Morandi, A.; Lagana, M.; Dalla Volta, A.; Bertagna, F.; et al. Treatment With Y-90/Lu-177-DOTATOC in Patients with Metastatic Adrenocortical Carcinoma Expressing Somatostatin Receptors. J. Clin. Endocr. Metab. 2020, 105, E1–E5. [Google Scholar] [CrossRef] [PubMed]
  36. Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021, 385, 1091–1103. [Google Scholar] [CrossRef] [PubMed]
  37. Crowley, M.J.; Scognamiglio, T.; Liu, Y.F.; Kleiman, D.A.; Beninato, T.; Aronova, A.; Liu, H.; Jhanwar, Y.S.; Molina, A.; Tagawa, S.T.; et al. Prostate-Specific Membrane Antigen Is a Potential Antiangiogenic Target in Adrenocortical Carcinoma. J. Clin. Endocrinol. Metab. 2016, 101, 981–987. [Google Scholar] [CrossRef]
  38. Arora, S.; Damle, N.A.; Aggarwal, S.; Passah, A.; Behera, A.; Arora, G.; Bal, C.; Tripathi, M. Prostate-Specific Membrane Antigen Expression in Adrenocortical Carcinoma on 68Ga-Prostate-Specific Membrane Antigen PET/CT. Clin. Nucl. Med. 2018, 43, 449–451. [Google Scholar] [CrossRef] [PubMed]
  39. Chatterjee, S.; Behnam Azad, B.; Nimmagadda, S. The intricate role of CXCR4 in cancer. Adv. Cancer Res. 2014, 124, 31–82. [Google Scholar] [CrossRef]
  40. Weiss, I.D.; Huff, L.M.; Evbuomwan, M.O.; Xu, X.; Dang, H.D.; Velez, D.S.; Singh, S.P.; Zhang, H.W.H.; Gardina, P.J.; Lee, J.H.; et al. Screening of cancer tissue arrays identifies CXCR4 on adrenocortical carcinoma: Correlates with expression and quantification on metastases using Cu-64-plerixafor PET. Oncotarget 2017, 8, 73387–73406. [Google Scholar] [CrossRef]
  41. Chifu, I.; Heinze, B.; Fuss, C.T.; Lang, K.A.; Kroiss, M.; Kircher, S.; Ronchi, C.L.; Altieri, B.; Schirbel, A.; Fassnacht, M.; et al. Impact of the Chemokine Receptors CXCR4 and CXCR7 on Clinical Outcome in Adrenocortical Carcinoma. Front. Endocrinol. 2020, 11, 597878. [Google Scholar] [CrossRef]
  42. Buck, A.K.; Haug, A.; Dreher, N.; Lambertini, A.; Higuchi, T.; Lapa, C.; Weich, A.; Pomper, M.G.; Wester, H.J.; Zehndner, A.; et al. Imaging of C-X-C Motif Chemokine Receptor 4 Expression in 690 Patients with Solid or Hematologic Neoplasms Using (68)Ga-Pentixafor PET. J. Nucl. Med. 2022, 63, 1687–1692. [Google Scholar] [CrossRef] [PubMed]
  43. Bluemel, C.; Hahner, S.; Heinze, B.; Fassnacht, M.; Kroiss, M.; Bley, T.A.; Wester, H.J.; Kropf, S.; Lapa, C.; Schirbel, A.; et al. Investigating the Chemokine Receptor 4 as Potential Theranostic Target in Adrenocortical Cancer Patients. Clin. Nucl. Med. 2017, 42, E29–E34. [Google Scholar] [CrossRef] [PubMed]
  44. Buck, A.K.; Serfling, S.E.; Lindner, T.; Hanscheid, H.; Schirbel, A.; Hahner, S.; Fassnacht, M.; Einsele, H.; Werner, R.A. CXCR4-targeted theranostics in oncology. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 4133–4144. [Google Scholar] [CrossRef] [PubMed]
  45. Weber, M.M.; Lang, J.; Abedinpour, F.; Zeilberger, K.; Adelmann, B.; Engelhardt, D. Different Inhibitory Effect of Etomidate and Ketoconazole on the Human Adrenal-Steroid Biosynthesis. Clin. Investig. 1993, 71, 933–938. [Google Scholar] [CrossRef] [PubMed]
  46. Damani, L.A.; Mitterhauser, M.; Zolle, I.; Lin, G.; Oehler, E.; Ho, Y.P. Metabolic and Pharmacokinetic Considerations in the Design of 2-Phenyl Substituted Metyrapone Derivatives—2-Methoxyphenylmetyrapone as a Radioligand for Functional Diagnosis of Adrenal Pathology. Nucl. Med. Biol. 1995, 22, 1067–1074. [Google Scholar] [CrossRef] [PubMed]
  47. Bergstrom, M.; Bonasera, T.A.; Lu, L.; Bergstrom, E.; Backlin, C.; Juhlin, C.; Langstrom, B. In vitro and in vivo primate evaluation of carbon-11-etomidate and carbon-11-metomidate as potential tracers for PET imaging of the adrenal cortex and its tumors. J. Nucl. Med. 1998, 39, 982–989. [Google Scholar] [PubMed]
  48. Bergstrom, M.; Juhlin, C.; Bonasera, T.A.; Sundin, A.; Rastad, J.; Akerstrom, G.; Langstrom, B. PET imaging of adrenal cortical tumors with the 11beta-hydroxylase tracer 11C-metomidate. J. Nucl. Med. 2000, 41, 275–282. [Google Scholar] [PubMed]
  49. Hennings, J.; Lindhe, O.; Bergstrom, M.; Langstrom, B.; Sundin, A.; Hellman, P. [(11)C]metomidate positron emission tomography of adrenocortical tumors in correlation with histopathological findings. J. Clin. Endocr. Metab. 2006, 91, 1410–1414. [Google Scholar] [CrossRef]
  50. Schirbel, A.; Zolle, I.; Hammerschmidt, F.; Berger, M.L.; Schiller, D.; Kvaternik, H.; Reiners, C. [I-123/131]Iodometomidate as a radioligand for functional diagnosis of adrenal disease: Synthesis, structural requirements and biodistribution. Radiochim. Acta 2004, 92, 297–303. [Google Scholar] [CrossRef]
  51. Hahner, S.; Stuermer, A.; Kreissl, M.; Reiners, C.; Fassnacht, M.; Haenscheid, H.; Beuschlein, F.; Zink, M.; Lang, K.; Allolio, B.; et al. [123I]Iodometomidate for molecular imaging of adrenocortical cytochrome P450 family 11B enzymes. J. Clin. Endocrinol. Metab. 2008, 93, 2358–2365. [Google Scholar] [CrossRef] [PubMed]
  52. Zolle, I.M.; Berger, M.L.; Hammerschmidt, F.; Hahner, S.; Schirbel, A.; Peric-Simov, B. New Selective Inhibitors of Steroid 11 beta-Hydroxylation in the Adrenal Cortex. Synthesis and Structure-Activity Relationship of Potent Etomidate Analogues (vol 51, pg 2244, 2008). J. Med. Chem. 2008, 51, 7652. [Google Scholar] [CrossRef]
  53. Hahner, S.; Kreissl, M.C.; Fassnacht, M.; Haenscheid, H.; Bock, S.; Verburg, F.A.; Knoedler, P.; Lang, K.; Reiners, C.; Buck, A.K.; et al. Functional Characterization of Adrenal Lesions Using [I-123] IMTO-SPECT/CT. J. Clin. Endocr. Metab. 2013, 98, 1508–1518. [Google Scholar] [CrossRef] [PubMed]
  54. Hahner, S.; Kreissl, M.C.; Fassnacht, M.; Haenscheid, H.; Knoedler, P.; Lang, K.; Buck, A.K.; Reiners, C.; Allolio, B.; Schirbel, A. [I-131]Iodometomidate for Targeted Radionuclide Therapy of Advanced Adrenocortical Carcinoma. J. Clin. Endocr. Metab. 2012, 97, 914–922. [Google Scholar] [CrossRef] [PubMed]
  55. Heinze, B.; Schirbel, A.; Nannen, L.; Michelmann, D.; Hartrampf, P.E.; Bluemel, C.; Schneider, M.; Herrmann, K.; Haenscheid, H.; Fassnacht, M.; et al. Novel CYP11B-ligand [I-123/131]IMAZA as promising theranostic tool for adrenocortical tumors: Comprehensive preclinical characterization and first clinical experience. Eur. J. Nucl. Med. Mol. I 2021, 49, 301–310. [Google Scholar] [CrossRef] [PubMed]
  56. Hahner, S.; Hartrampf, P.E.; Mihatsch, P.W.; Nauerz, M.; Heinze, B.; Hanscheid, H.; Fuss, C.T.; Werner, R.A.; Pamporaki, C.; Kroiss, M.; et al. Targeting 11-Beta Hydroxylase With [I-131]IMAZA: A Novel Approach for the Treatment of Advanced Adrenocortical Carcinoma. J. Clin. Endocr. Metab. 2022, 107, E1348–E1355. [Google Scholar] [CrossRef] [PubMed]
  57. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef]
  58. Aslani, A.; Snowdon, G.M.; Bailey, D.L.; Schembri, G.P.; Bailey, E.A.; Pavlakis, N.; Roach, P.J. Lutetium-177 DOTATATE Production with an Automated Radiopharmaceutical Synthesis System. Asia Ocean. J. Nucl. Med. Biol. 2015, 3, 107–115. [Google Scholar]
  59. Schottelius, M.; Osl, T.; Poschenrieder, A.; Hoffmann, F.; Beykan, S.; Hanscheid, H.; Schirbel, A.; Buck, A.K.; Kropf, S.; Schwaiger, M.; et al. [(177)Lu]pentixather: Comprehensive Preclinical Characterization of a First CXCR4-directed Endoradiotherapeutic Agent. Theranostics 2017, 7, 2350–2362. [Google Scholar] [CrossRef]
  60. Hanscheid, H.; Schirbel, A.; Hartrampf, P.; Kraus, S.; Werner, R.A.; Einsele, H.; Wester, H.J.; Lassmann, M.; Kortum, M.; Buck, A.K. Biokinetics and Dosimetry of (177)Lu-Pentixather. J. Nucl. Med. 2022, 63, 754–760. [Google Scholar] [CrossRef]
  61. Wichmann, C.W.; Ackermann, U.; Poniger, S.; Young, K.; Nguyen, B.; Chan, G.; Sachinidis, J.; Scott, A.M. Automated radiosynthesis of [(68) Ga]Ga-PSMA-11 and [(177) Lu]Lu-PSMA-617 on the iPHASE MultiSyn module for clinical applications. J. Label. Comp. Radiopharm. 2021, 64, 140–146. [Google Scholar] [CrossRef]
Figure 1. [131I]IMAZA therapy in a 53-year-old patient with metastatic adrenocortical cancer. FDG PET maximum intensity projection (MIP) is shown at baseline (A). Post-therapeutic whole-body scintigraphy 2 days after first therapy (B) shows concordant tracer accumulation to FDG PET/CT. Response assessment after 3 and 8 months (C,D) shows a significant decrease in metabolic activity and a reduction in the diameter of the target lesion of 26%. After a progression-free survival of 18 months, a second therapy with [131I]IMAZA was applied. The patient died after an overall survival of 56 months after the first [131I]IMAZA therapy.
Figure 1. [131I]IMAZA therapy in a 53-year-old patient with metastatic adrenocortical cancer. FDG PET maximum intensity projection (MIP) is shown at baseline (A). Post-therapeutic whole-body scintigraphy 2 days after first therapy (B) shows concordant tracer accumulation to FDG PET/CT. Response assessment after 3 and 8 months (C,D) shows a significant decrease in metabolic activity and a reduction in the diameter of the target lesion of 26%. After a progression-free survival of 18 months, a second therapy with [131I]IMAZA was applied. The patient died after an overall survival of 56 months after the first [131I]IMAZA therapy.
Pharmaceuticals 17 00025 g001
Figure 2. Schematic depiction of possible theranostic targets in adrenocortical carcinoma (black font) and the respective therapeutic radiopharmaceuticals (exemplary, blue font).
Figure 2. Schematic depiction of possible theranostic targets in adrenocortical carcinoma (black font) and the respective therapeutic radiopharmaceuticals (exemplary, blue font).
Pharmaceuticals 17 00025 g002
Figure 3. Chemical structure of possible theranostic radiopharmaceuticals for treatment of ACC. To date, [131I]IMAZA is the only compound that has been already used in patients.
Figure 3. Chemical structure of possible theranostic radiopharmaceuticals for treatment of ACC. To date, [131I]IMAZA is the only compound that has been already used in patients.
Pharmaceuticals 17 00025 g003
Figure 4. Photo and scheme of module for radiosynthesis of [131I]IMAZA.
Figure 4. Photo and scheme of module for radiosynthesis of [131I]IMAZA.
Pharmaceuticals 17 00025 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Michalski, K.; Schlötelburg, W.; Hartrampf, P.E.; Kosmala, A.; Buck, A.K.; Hahner, S.; Schirbel, A. Radiopharmaceuticals for Treatment of Adrenocortical Carcinoma. Pharmaceuticals 2024, 17, 25. https://doi.org/10.3390/ph17010025

AMA Style

Michalski K, Schlötelburg W, Hartrampf PE, Kosmala A, Buck AK, Hahner S, Schirbel A. Radiopharmaceuticals for Treatment of Adrenocortical Carcinoma. Pharmaceuticals. 2024; 17(1):25. https://doi.org/10.3390/ph17010025

Chicago/Turabian Style

Michalski, Kerstin, Wiebke Schlötelburg, Philipp E. Hartrampf, Aleksander Kosmala, Andreas K. Buck, Stefanie Hahner, and Andreas Schirbel. 2024. "Radiopharmaceuticals for Treatment of Adrenocortical Carcinoma" Pharmaceuticals 17, no. 1: 25. https://doi.org/10.3390/ph17010025

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop