Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success?
Abstract
:Simple Summary
Abstract
1. Introduction
2. Tumor Immune Microenvironment in Adrenocortical Carcinoma
2.1. Immune Cells
2.2. Cancer Cells
2.2.1. Altered MHC II Expression
2.2.2. TLR4 and CD14 Expression
2.2.3. CD276 (B7-H3) Expression
2.3. Adipose Stem Cells
2.4. Immunosuppressive Role of Glucocorticoids on TME
2.5. Locally Produced Androgens
2.6. Alteration of Oncogenic Pathways
3. Biomarkers of Immunotherapy in ACC
3.1. PD-L1
3.2. TMB
3.3. MSI/MMR
3.4. Other Biomarkers
4. Immunotherapy Trials in ACC
4.1. Clinical Trials with Immune Checkpoint Inhibitors
4.2. Other Immunotherapeutic Approaches
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
ACC | adrenocortical carcinoma |
APC | antigen presenting cells |
ASC | adipose stem cells |
CAF | cancer associated fibroblasts |
CoC I | cluster of cluster I |
DAMPs | damage associated molecular patterns |
DHEA | Dehydroepiandrosterone |
DHEA-S | dehydroepiandrosterone sulfate |
dMMR | defective mismatch repair |
ICI | immune checkpoint inhibitors |
IO | Immunotherapy |
LFS | Li Fraumeni syndrome |
LPS | Lipopolysaccharide |
MART-1 | melanoma antigen recognized by T-cells |
MD2 | myeloid differentiation factor 2 |
MSI | microsatellite instability |
MSS | microsatellite stable |
NF-κΒ | nuclear factor κ-light-chain-enhancer of activated B cells |
OS | overall survival |
PD | progressive disease |
PD-1 | programmed cell death protein-1 |
PDL-1 | programmed cell death ligand-1 |
PFS | progression free survival |
RFS | recurrence-free survival |
SD | stable disease |
StAR | steroidogenic acute regulatory protein |
TCR | T-cell receptor |
TIME | tumor immune microenvironment |
TILs | tumor infiltrating lymphocytes |
TLR | Toll-like receptor |
TMB | tumor mutation burden |
TME | tumor microenvironment |
TNF-α | tumor necrosis factor-α |
Treg | T regulatory cells |
VEGF | vascular endothelial growth factor |
References
- WHO. Classification of Tumors of Endocrine Organs WHO/IARC Classification of Tumors, 4th ed.; Lloyd, R.V., Osamura, R.Y., Kloppel, G., Rosai, J., Eds.; WHO: Geneva, Switzerland, 2017; Volume 10. [Google Scholar]
- Berruti, A.; Grisanti, S.; Pulzer, A.; Claps, M.; Daffara, F.; Loli, P.; Mannelli, M.; Boscaro, M.; Arvat, E.; Tiberio, G.; et al. Long-Term Outcomes of Adjuvant Mitotane Therapy in Patients with Radically Resected Adrenocortical Carcinoma. J. Clin. Endocrinol. Metab. 2017, 102, 1358–1365. [Google Scholar] [CrossRef] [PubMed]
- Berruti, A.; Terzolo, M.; Sperone, P.; Pia, A.; Della Casa, S.; Gross, D.J.; Carnaghi, C.; Casali, P.; Porpiglia, F.; Mantero, F.; et al. Etoposide, doxorubicin and cisplatin plus mitotane in the treatment of advanced adrenocortical carcinoma: A large prospective phase II trial. Endocr. Relat. Cancer 2005, 12, 657–666. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Megerle, F.; Herrmann, W.; Schloetelburg, W.; Ronchi, C.L.; Pulzer, A.; Quinkler, M.; Beuschlein, F.; Hahner, S.; Kroiss, M.; Fassnacht, M.; et al. Mitotane Monotherapy in Patients With Advanced Adrenocortical Carcinoma. J. Clin. Endocrinol. Metab. 2018, 103, 1686–1695. [Google Scholar] [CrossRef] [PubMed]
- Henning, J.E.K.; Deutschbein, T.; Altieri, B.; Steinhauer, S.; Kircher, S.; Sbiera, S.; Wild, V.; Schlötelburg, W.; Kroiss, M.; Perotti, P.; et al. Gemcitabine-Based Chemotherapy in Adrenocortical Carcinoma: A Multicenter Study of Efficacy and Predictive Factors. J. Clin. Endocrinol. Metab. 2017, 102, 4323–4332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fassnacht, M. European Journal of Endocrinology European Society of Endocrinology Clinical Practice Guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Net-work for the Study of Adrenal Tumors. Eur. J. Endocrinol. 2018, 179, G1–G46. [Google Scholar] [CrossRef]
- Ribas, A.; Hamid, O.; Daud, A.; Hodi, F.S.; Wolchok, J.D.; Kefford, R.; Joshua, A.M.; Patnaik, A.; Hwu, W.-J.; Weber, J.S.; et al. Association of Pembrolizumab With Tumor Response and Survival Among Patients with Advanced Melanoma. JAMA 2016, 315, 1600–1609. [Google Scholar] [CrossRef]
- Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef]
- Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Procopio, G.; Plimack, E.R.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1803–1813. [Google Scholar] [CrossRef]
- Cohen, E.E.W.; Bell, R.B.; Bifulco, C.B.; Burtness, B.; Gillison, M.L.; Harrington, K.J.; Le, Q.-T.; Lee, N.Y.; Leidner, R.; Lewis, R.L.; et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J. Immunother. Cancer 2019, 7, 184. [Google Scholar] [CrossRef] [Green Version]
- Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef]
- Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef] [Green Version]
- Denkert, C.; Von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; Budczies, J.; Huober, J.; Klauschen, F.; Furlanetto, J.; et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: A pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018, 19, 40–50. [Google Scholar] [CrossRef]
- Fu, Q.; Chen, N.; Ge, C.; Li, R.; Li, Z.; Zeng, B.; Li, C.; Wang, Y.; Xue, Y.; Song, X.; et al. Prognostic value of tumor-infiltrating lymphocytes in melanoma: A systematic review and meta-analysis. OncoImmunology 2019, 8, e1593806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paré, L.; Pascual, T.; Seguí, E.; Teixidó, C.; Gonzalez-Cao, M.; Galván, P.; Rodríguez, A.; González, B.; Cuatrecasas, M.; Pineda, E.; et al. Association between PD1 mRNA and response to anti-PD1 monotherapy across multiple cancer types. Ann. Oncol. 2018, 29, 2121–2128. [Google Scholar] [CrossRef] [PubMed]
- Thorsson, V.; Gibbs, D.L.; Brown, S.; Wolf, D.; Bortone, D.S.; Ouyang, T.-H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The Immune Landscape of Cancer. Immunity 2018, 48, 812–830.e14. [Google Scholar] [CrossRef] [Green Version]
- Mohan, D.R.; Lerario, A.M.; Hammer, G.D. Therapeutic Targets for Adrenocortical Carcinoma in the Genomics Era. J. Endocr. Soc. 2018, 2, 1259–1274. [Google Scholar] [CrossRef]
- Peng, Y.; Song, Y.; Ding, J.; Li, N.; Zhang, Z.; Wang, H. Identification of immune-related biomarkers in adrenocortical carcinoma. Int. Immunopharmacol. 2020, 88, 106930. [Google Scholar] [CrossRef]
- Landwehr, L.-S.; 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 2019, 8, e000469. [Google Scholar] [CrossRef]
- Tian, X.; Xu, W.; Wang, Y.; Anwaier, A.; Wang, H.; Wan, F.; Zhu, Y.; Cao, D.; Shi, G.; Zhu, Y.; et al. Identification of tumor-infiltrating immune cells and prognostic validation of tumor-infiltrating mast cells in adrenocortical carcinoma: Results from bioinformatics and real-world data. OncoImmunology 2020, 9, 1784529. [Google Scholar] [CrossRef]
- Parise, I.Z.S.; Parise, G.A.; Noronha, L.; Surakhy, M.; Woiski, T.D.; Silva, D.B.; Costa, T.E.-J.B.; Del-Valle, M.H.C.P.; Komechen, H.; Rosati, R.; et al. The Prognostic Role of CD8+ T Lymphocytes in Childhood Adrenocortical Carcinomas Compared to Ki-67, PD-1, PD-L1, and the Weiss Score. Cancers 2019, 11, 1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bagante, F.; Tran, T.B.; Postlewait, L.M.; Maithel, S.K.; Wang, T.S.; Evans, D.B.; Hatzaras, I.; Shenoy, R.; Phay, J.E.; Keplinger, K.; et al. Neutrophil-lymphocyte and platelet-lymphocyte ratio as predictors of disease specific survival after resection of adrenocortical carcinoma. J. Surg. Oncol. 2015, 112, 164–172. [Google Scholar] [CrossRef] [Green Version]
- Jackson, R.; McNicol, A.M.; Farquharson, M.; Foulis, A.K. Class II MHC expression in normal adrenal cortex and cortical cells in autoimmune Addison’s disease. J. Pathol. 1988, 155, 113–120. [Google Scholar] [CrossRef] [PubMed]
- Wolkersdörfer, G.W.; Marx, C.; Brown, J.; Schröder, S.; Füssel, M.; Rieber, E.P.; Kuhlisch, E.; Ehninger, G.; Bornstein, S.R. Prevalence of HLA-DRB1 Genotype and Altered Fas/Fas Ligand Expression in Adrenocortical Carcinoma. J. Clin. Endocrinol. Metab. 2005, 90, 1768–1774. [Google Scholar] [CrossRef] [Green Version]
- Hahne, M.; Rimoldi, D.; Schroter, M.; Romero, P.; Schreier, M.; French, L.E.; Schneider, P.; Bornand, T.; Fontana, A.; Lienard, D.; et al. Melanoma Cell Expression of Fas(Apo-1/CD95) Ligand: Implications for Tumor Immune Escape. Science 1996, 274, 1363–1366. [Google Scholar] [CrossRef]
- Xiao, W.; Ibrahim, M.L.; Redd, P.S.; Klement, J.D.; Lu, C.; Yang, D.; Savage, N.M.; Liu, K. Loss of Fas Expression and Function Is Coupled with Colon Cancer Resistance to Immune Checkpoint Inhibitor Immunotherapy. Mol. Cancer Res. 2019, 17, 420–430. [Google Scholar] [CrossRef] [Green Version]
- Shibakita, M.; Tachibana, M.; Dhar, D.K.; Kotoh, T.; Kinugasa, S.; Kubota, H.; Nagasue, N. Prognostic Significance of Fas and Fas Ligand Expressions in Human Esophageal Cancer. Clin. Cancer Res. 1999, 5, 9. [Google Scholar]
- Pinto, E.M.; Rodriguez-Galindo, C.; Choi, J.K.; Pounds, S.; Liu, Z.; Neale, G.; Finkelstein, D.; Hicks, J.M.; Pappo, A.S.; Figueiredo, B.C.; et al. Prognostic Significance of Major Histocompatibility Complex Class II Expression in Pediatric Adrenocortical Tumors: A St. Jude and Children’s Oncology Group Study. Clin. Cancer Res. 2016, 22, 6247–6255. [Google Scholar] [CrossRef] [Green Version]
- Shcheblyakov, D.; Logunov, D.; Tukhvatulin, A.; Shmarov, M.; Naroditsky, B.; Ginzburg, A.L. Toll-Like Receptors (TLRs): The Role in Tumor Progression. Acta Naturae 2010, 2, 21–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Urban-Wojciuk, Z.; Khan, M.M.; Oyler, B.L.; Fåhraeus, R.; Marek-Trzonkowska, N.; Nita-Lazar, A.; Hupp, T.R.; Goodlett, D.R. The Role of TLRs in Anti-cancer Immunity and Tumor Rejection. Front. Immunol. 2019, 10, 2388. [Google Scholar] [CrossRef] [PubMed]
- Matsumoto, M.; Takeda, Y.; Tatematsu, M.; Seya, T. Toll-Like Receptor 3 Signal in Dendritic Cells Benefits Cancer Immunotherapy. Front. Immunol. 2017, 8, 1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanczkowski, W.; Tymoszuk, P.; Ehrhart-Bornstein, M.; Wirth, M.P.; Zacharowski, K.; Bornstein, S.R. Abrogation of TLR4 and CD14 Expression and Signaling in Human Adrenocortical Tumors. J. Clin. Endocrinol. Metab. 2010, 95, 421–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, P.; Xiong, Y.; Yue, J.; Hanley, S.J.B.; Watari, H. B7H3 As a Promoter of Metastasis and Promising Therapeutic Target. Front. Oncol. 2018, 8, 264. [Google Scholar] [CrossRef] [Green Version]
- Picarda, E.; Ohaegbulam, K.C.; Zang, X. Molecular Pathways: Targeting B7-H3 (CD276) for Human Cancer Immunotherapy. Clin. Cancer Res. 2016, 22, 3425–3431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, J.; Liu, Z.; Pei, T.; Xiao, Y.; Zhou, L.; Tang, Y.; Zhou, C.; Wu, K.; Zhang, F.; Zhang, F.; et al. Clinicopathological and Prognostic Characteristics of CD276 (B7-H3) Expression in Adrenocortical Carcinoma. Dis. Markers 2020, 2020, 1–10. [Google Scholar] [CrossRef]
- Song, Y.H.; Shon, S.H.; Shan, M.; Stroock, A.D.; Fischbachtreschl, C. Adipose-derived stem cells increase angiogenesis through matrix metalloproteinase-dependent collagen remodeling. Integr. Biol. 2016, 8, 205–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scioli, M.G.; Storti, G.; D’Amico, F.; Gentile, P.; Kim, B.-S.; Cervelli, V.; Orlandi, A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int. J. Mol. Sci. 2019, 20, 3296. [Google Scholar] [CrossRef] [Green Version]
- Armignacco, R.; Cantini, G.; Poli, G.; Guasti, D.; Nesi, G.; Romagnoli, P.; Mannelli, M.; Luconi, M. The Adipose Stem Cell as a Novel Metabolic Actor in Adrenocortical Carcinoma Progression: Evidence from an In Vitro Tumor Microenvironment Crosstalk Model. Cancers 2019, 11, 1931. [Google Scholar] [CrossRef] [Green Version]
- Strong, A.L.; Ohlstein, J.F.; Biagas, B.A.; Rhodes, L.V.; Pei, D.T.; Tucker, H.A.; Llamas, C.; Bowles, A.C.; DuTreil, M.F.; Zhang, S.; et al. Leptin produced by obese adipose stromal/stem cells enhances proliferation and metastasis of estrogen receptor positive breast cancers. Breast Cancer Res. 2015, 17, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.; Chang, Y.-C.; Lan, M.S.; Breslin, M. Leptin stimulates ovarian cancer cell growth and inhibits apoptosis by increasing cyclin D1 and Mcl-1 expression via the activation of the MEK/ERK1/2 and PI3K/Akt signaling pathways. Int. J. Oncol. 2013, 42, 1113–1119. [Google Scholar] [CrossRef] [Green Version]
- Fiorentini, C.; Grisanti, S.; Cosentini, D.; Abate, A.; Rossini, E.; Berruti, A.; Sigala, S. Molecular Drivers of Potential Immunotherapy Failure in Adrenocortical Carcinoma. J. Oncol. 2019, 2019, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Zheng, S.; Cherniack, A.D.; Dewal, N.; Moffitt, R.A.; Danilova, L.; Murray, B.A.; Lerario, A.M.; Else, T.; Knijnenburg, T.A.; Ciriello, G.; et al. Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma. Cancer Cell 2016, 29, 723–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coutinho, A.E.; Chapman, K.E. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2–13. [Google Scholar] [CrossRef] [PubMed]
- Guendisch, S.; Boeckeler, E.; Behrends, U.; Amtmann, E.; Ehrhardt, H.; Jeremias, I. Gluco-corticoids Augment Survival and Proliferation of Tumor Cells. Anticancer Res. 2012, 32, 10. [Google Scholar]
- Celada, A.; McKercher, S.; Maki, R. Repression of major histocompatibility complex IA expression by glucocorticoids: The glucocorticoid receptor inhibits the DNA binding of the X box DNA binding protein. J. Exp. Med. 1993, 177, 691–698. [Google Scholar] [CrossRef]
- Curtale, G.; Renzi, T.A.; Drufuca, L.; Rubino, M.; Locati, M. Glucocorticoids downregulate TLR4 signaling activity via its direct targeting by miR-511-5p. Eur. J. Immunol. 2017, 47, 2080–2089. [Google Scholar] [CrossRef]
- Casey, R.; Giger, O.; Seetho, I.; Marker, A.; Pitfield, D.; Boyle, L.; Gurnell, M.; Shaw, A.; Tischkowitz, M.; Maher, E.; et al. Rapid disease progression in a patient with mismatch repair-deficient and cortisol secreting adrenocortical carcinoma treated with pembrolizumab. Semin. Oncol. 2018, 45, 151–155. [Google Scholar] [CrossRef]
- Caccese, M.; Barbot, M.; Ceccato, F.; Padovan, M.; Gardiman, M.P.; Fassan, M.; Denaro, L.; Emanuelli, E.; D’Avella, D.; Scaroni, C.; et al. Rapid disease progression in patient with mismatch-repair deficiency pituitary ACTH-secreting adenoma treated with checkpoint inhibitor pembrolizumab. Anti-Cancer Drugs 2020, 31, 199–204. [Google Scholar] [CrossRef]
- Daniel, E.; Aylwin, S.; Mustafa, O.; Ball, S.; Munir, A.; Boelaert, K.; Chortis, V.; Cuthbertson, D.J.; Daousi, C.; Rajeev, S.P.; et al. Effectiveness of Metyrapone in Treating Cushing’s Syndrome: A Retrospective Multicenter Study in 195 Patients. J. Clin. Endocrinol. Metab. 2015, 100, 4146–4154. [Google Scholar] [CrossRef] [PubMed]
- Fleseriu, M.; Biller, B.M.K.; Findling, J.W.; Molitch, M.E.; Schteingart, D.E.; Gross, C.; Auchus, R.; Bailey, T.; Carroll, T.; Colleran, K.; et al. Mifepristone, a Glucocorticoid Receptor Antagonist, Produces Clinical and Metabolic Benefits in Patients with Cushing’s Syndrome. J. Clin. Endocrinol. Metab. 2012, 97, 2039–2049. [Google Scholar] [CrossRef] [Green Version]
- Castinetti, F.; Fassnacht, M.; Johanssen, S.; Terzolo, M.; Bouchard, P.; Chanson, P.; Cao, C.D.; Morange, I.; Pico, A.; Ouzounian, S.; et al. Merits and pitfalls of mifepristone in Cushing’s syndrome. Eur. J. Endocrinol. 2009, 160, 1003–1010. [Google Scholar] [CrossRef] [Green Version]
- Tagawa, N.; Minamitani, E.; Yamaguchi, Y.; Kobayashi, Y. Alternative mechanism for anti-obesity effect of dehydroepiandrosterone: Possible contribution of 11β-hydroxysteroid dehydrogenase type 1 inhibition in rodent adipose tissue. Steroids 2011, 76, 1546–1553. [Google Scholar] [CrossRef] [PubMed]
- Khorram, O.; Vu, L.; Yen, S.S.C. Activation of Immune Function by Dehydroepiandrosterone (DHEA) in Age-Advanced Men. Journals Gerontol. Ser. A Boil. Sci. Med Sci. 1997, 52, M1–M7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Opposing Effects of Dehydroepiandrosterone and Dexamethasone on the Generation of Monocyte-Derived Dendritic Cells. Eur. J. Endocrinol. 2000, 143. Available online: https://eje.bioscientifica.com/view/journals/eje/143/5/687.xml (accessed on 8 February 2021).
- Straub, R.H.; Konečná, L.; Hrach, S.; Rothe, G.; Kreutz, M.; Scholmerich, J.; Falk, W.; Lang, B. Serum Dehydroepiandrosterone (DHEA) and DHEA Sulfate Are Negatively Correlated with Serum Interleukin-6 (IL-6), and DHEA Inhibits IL-6 Secretion from Mononuclear Cells in Manin Vitro: Possible Link between Endocrinosenescence and Immunosenescence. J. Clin. Endocrinol. Metab. 1998, 83, 2012–2017. [Google Scholar] [CrossRef] [PubMed]
- Spranger, S.; Gajewski, T.F. Mechanisms of Tumor Cell–Intrinsic Immune Evasion. Annu. Rev. Cancer Biol. 2018, 2, 213–228. [Google Scholar] [CrossRef]
- Jiménez-Sánchez, A.; Memon, D.; Pourpe, S.; Veeraraghavan, H.; Li, Y.; Vargas, H.A.; Gill, M.B.; Park, K.J.; Zivanovic, O.; Konner, J.; et al. Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient. Cell 2017, 170, 927–938.e20. [Google Scholar] [CrossRef] [Green Version]
- Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Patel, L.; Mills, G.B.; Lu, K.H.; Sood, A.K.; Ding, L.; Kucherlapati, R.; Mardis, E.R.; Levine, D.A.; Shmulevich, I.; et al. Clinical Significance of CTNNB1 Mutation and Wnt Pathway Activation in Endometrioid Endometrial Carcinoma. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef] [Green Version]
- Krishnamurthy, N.; Kurzrock, R. Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat. Rev. 2018, 62, 50–60. [Google Scholar] [CrossRef]
- Diamond, J.R.; Becerra, C.; Richards, D.; Mita, A.; Osborne, C.; O’Shaughnessy, J.; Zhang, C.; Henner, R.; Kapoun, A.M.; Xu, L.; et al. Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer. Breast Cancer Res. Treat. 2020, 184, 53–62. [Google Scholar] [CrossRef]
- Davis, S.L.; Cardin, D.B.; Shahda, S.; Lenz, H.-J.; Dotan, E.; O’Neil, B.H.; Kapoun, A.M.; Stagg, R.J.; Berlin, J.; Messersmith, W.A.; et al. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer. Investig. New Drugs 2020, 38, 821–830. [Google Scholar] [CrossRef] [Green Version]
- Ko, A.H.; Chiorean, E.G.; Kwak, E.L.; Lenz, H.-J.; Nadler, P.I.; Wood, D.L.; Fujimori, M.; Inada, T.; Kouji, H.; McWilliams, R.R. Final results of a phase Ib dose-escalation study of PRI-724, a CBP/beta-catenin modulator, plus gemcitabine (GEM) in patients with advanced pancreatic adenocarcinoma (APC) as second-line therapy after FOLFIRINOX or FOLFOX. J. Clin. Oncol. 2016, 34, e15721. [Google Scholar] [CrossRef]
- Gaujoux, S.; Hantel, C.; Launay, P.; Bonnet, S.; Perlemoine, K.; Lefèvre, L.; Guillaud-Bataille, M.; Beuschlein, F.; Tissier, F.; Bertherat, J.; et al. Silencing Mutated β-Catenin Inhibits Cell Proliferation and Stimulates Apoptosis in the Adrenocortical Cancer Cell Line H295R. PLoS ONE 2013, 8, e55743. [Google Scholar] [CrossRef]
- Leal, L.F.; Bueno, A.C.; Gomes, D.C.; Abduch, R.; De Castro, M.; Antonini, S.R. Inhibition of the Tcf/beta-catenin complex increases apoptosis and impairs adrenocortical tumor cell proliferation and adrenal steroidogenesis. Oncotarget 2015, 6, 43016–43032. [Google Scholar] [CrossRef]
- Ross, J.S.; Wang, K.; Rand, J.V.; Gay, L.; Presta, M.J.; Sheehan, C.E.; Ali, S.M.; Elvin, J.A.; Labrecque, E.; Hiemstra, C.; et al. Next-generation sequencing of adrenocortical carcinoma reveals new routes to targeted therapies. J. Clin. Pathol. 2014, 67, 968–973. [Google Scholar] [CrossRef] [Green Version]
- Vatrano, S.; Volante, M.; Duregon, E.; Giorcelli, J.; Izzo, S.; Rapa, I.; Votta, A.; Germano, A.; Scagliotti, G.; Berruti, A.; et al. Detailed genomic characterization identifies high heterogeneity and histotype-specific genomic profiles in adrenocortical carcinomas. Mod. Pathol. 2018, 31, 1257–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mantovani, F.; Walerych, D.; Del Sal, G. Targeting mutant p53 in cancer: A long road to precision therapy. FEBS J. 2016, 284, 837–850. [Google Scholar] [CrossRef] [PubMed]
- Wasserman, J.D.; Zambetti, G.P.; Malkin, D. Towards an understanding of the role of p53 in adrenocortical carcinogenesis. Mol. Cell. Endocrinol. 2012, 351, 101–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blagih, J.; Zani, F.; Chakravarty, P.; Hennequart, M.; Pilley, S.; Hobor, S.; Hock, A.K.; Walton, J.B.; Morton, J.P.; Gronroos, E.; et al. Cancer-Specific Loss of p53 Leads to a Modulation of Myeloid and T Cell Responses. Cell Rep. 2020, 30, 481–496.e6. [Google Scholar] [CrossRef]
- Long, J.; Wang, A.; Bai, Y.; Lin, J.; Yang, X.; Wang, D.; Yang, X.; Jiang, Y.; Zhao, H. Development and validation of a TP53-associated immune prognostic model for hepatocellular carcinoma. EBioMedicine 2019, 42, 363–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, G.; Cui, Y. New perspective on targeting the tumor suppressor p53 pathway in the tumor microenvironment to enhance the efficacy of immunotherapy. J. Immunother. Cancer 2015, 3, 9. [Google Scholar] [CrossRef] [Green Version]
- Altieri, B.; Ronchi, C.L.; Kroiss, M.; Fassnacht, M. Next-generation therapies for adrenocortical carcinoma. Best Pr. Res. Clin. Endocrinol. Metab. 2020, 34, 101434. [Google Scholar] [CrossRef] [PubMed]
- Jin, Y.; Tymen, S.D.; Chen, D.; Fang, Z.J.; Zhao, Y.; Dragas, D.; Dai, Y.; Marucha, P.T.; Zhou, X. MicroRNA-99 Family Targets AKT/mTOR Signaling Pathway in Dermal Wound Healing. PLoS ONE 2013, 8, e64434. [Google Scholar] [CrossRef]
- Conciatori, F.; Ciuffreda, L.; Bazzichetto, C.; Falcone, I.; Pilotto, S.; Bria, E.; Cognetti, F.; Milella, M. mTOR Cross-Talk in Cancer and Potential for Combination Therapy. Cancers 2018, 10, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Martino, M.C.; Van Koetsveld, P.M.; Feelders, R.A.; De Herder, W.W.; Dogan, F.; Janssen, J.A.M.J.L.; Bruinink, D.H.O.; Pivonello, C.; Waaijers, A.M.; Colao, A.; et al. IGF and mTOR pathway expression and in vitro effects of linsitinib and mTOR inhibitors in adrenocortical cancer. Endocrine 2019, 64, 673–684. [Google Scholar] [CrossRef] [Green Version]
- Doghman, M.; El Wakil, A.; Cardinaud, B.; Thomas, E.; Wang, J.; Zhao, W.; Valle, M.H.C.P.-D.; Figueiredo, B.C.; Zambetti, G.P.; Lalli, E. Regulation of Insulin-like Growth Factor–Mammalian Target of Rapamycin Signaling by MicroRNA in Childhood Adrenocortical Tumors. Cancer Res. 2010, 70, 4666–4675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fraenkel, M.; Gueorguiev, M.; Barak, D.; Salmon, A.; Grossman, A.B.; Gross, D.J. Everolimus therapy for progressive adrenocortical cancer. Endocrine 2013, 44, 187–192. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 Pathway: Current Researches in Cancer. Am. J. Cancer Res. 2020, 10, 727–742. Available online: http://www.ncbi.nlm.nih.gov/pubmed/32266087 (accessed on 7 February 2021). [PubMed]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti–PD-L1 Antibody in Patients with Advanced Cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gou, Q.; Dong, C.; Xu, H.; Khan, B.; Jin, J.; Liu, Q.; Shi, J.; Hou, Y. PD-L1 degradation pathway and immunotherapy for cancer. Cell Death Dis. 2020, 11, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Fay, A.P.; Signoretti, S.; Callea, M.; TelόG, H.; McKay, R.R.; Song, J.; Carvo, I.; Lampron, M.E.; Kaymakcalan, M.D.; Poli-De-Figueiredo, C.; et al. Programmed death ligand-1 expression in adrenocortical carcinoma: An exploratory biomarker study. J. Immunother. Cancer 2015, 3, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Billon, E.; Finetti, P.; Bertucci, A.; Niccoli, P.; Birnbaum, D.; Mamessier, E.; Bertucci, F. PDL1 expression is associated with longer postoperative, survival in adrenocortical carcinoma. OncoImmunology 2019, 8, e1655362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
- Colli, L.M.M.; Machiela, M.J.; Myers, T.A.; Jessop, L.; Yu, K.; Chanock, S.J. Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res. 2016, 76, 3767–3772. [Google Scholar] [CrossRef] [Green Version]
- Gara, S.K.; Lack, J.; Zhang, L.; Harris, E.; Cam, M.; Kebebew, E. Metastatic adrenocortical carcinoma displays higher mutation rate and tumor heterogeneity than primary tumors. Nat. Commun. 2018, 9, 1–11. [Google Scholar] [CrossRef]
- Bonneville, R.; Krook, M.A.; Kautto, E.A.; Miya, J.; Wing, M.R.; Chen, H.-Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of Microsatellite Instability Across 39 Cancer Types. JCO Precis. Oncol. 2017, 1, 1–15. [Google Scholar] [CrossRef]
- Mota, J.M.; Sousa, L.G.; Braghiroli, M.I.; Siqueira, L.T.; Neto, J.E.B.; Chapchap, P.; Hoff, A.A.D.O.; Hoff, P.M. Pembrolizumab for metastatic adrenocortical carcinoma with high mutational burden. Medicine 2018, 97, e13517. [Google Scholar] [CrossRef]
- Ruiz-Bañobre, J.; Kandimalla, R.; Goel, A. Predictive Biomarkers in Metastatic Colorectal Cancer: A Systematic Review. JCO Precis. Oncol. 2019, 3, 1–17. [Google Scholar] [CrossRef]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.; Kemberling, H.; Eyring, A.; Azad, N.S.; Laheru, D.; Donehower, R.C.; Crocenzi, T.S.; et al. Programmed death-1 blockade in mismatch repair deficient colorectal cancer. J. Clin. Oncol. 2016, 34, 103. [Google Scholar] [CrossRef]
- Duffy, M.J.; Crown, J. Biomarkers for Predicting Response to Immunotherapy with Immune Checkpoint Inhibitors in Cancer Patients. Clin. Chem. 2019, 65, 1228–1238. [Google Scholar] [CrossRef] [PubMed]
- Challis, B.G.; Kandasamy, N.; Powlson, A.S.; Koulouri, O.; Annamalai, A.K.; Happerfield, L.; Marker, A.J.; Arends, M.J.; Nik-Zainal, S.; Gurnell, M. Familial Adrenocortical Carcinoma in Association With Lynch Syndrome. J. Clin. Endocrinol. Metab. 2016, 101, 2269–2272. [Google Scholar] [CrossRef] [Green Version]
- Wright, J.P.; Montgomery, K.W.; Tierney, J.; Gilbert, J.; Solórzano, C.C.; Idrees, K. Ectopic, retroperitoneal adrenocortical carcinoma in the setting of Lynch syndrome. Fam. Cancer 2017, 17, 381–385. [Google Scholar] [CrossRef] [PubMed]
- Kaur, R.J.; Pichurin, P.N.; Hines, J.M.; Singh, R.J.; Grebe, S.K.; Bancos, I. Adrenal Cortical Carcinoma Associated With Lynch Syndrome: A Case Report and Review of Literature. J. Endocr. Soc. 2019, 3, 784–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raymond, V.M.; Everett, J.N.; Furtado, L.V.; Gustafson, S.L.; Jungbluth, C.R.; Gruber, S.B.; Hammer, G.D.; Stoffel, E.M.; Greenson, J.K.; Giordano, T.J.; et al. Adrenocortical Carcinoma Is a Lynch Syndrome–Associated Cancer. J. Clin. Oncol. 2013, 31, 3012–3018. [Google Scholar] [CrossRef]
- Busam, K.J.; Iversen, K.; Coplan, K.A.; Old, L.J.; Stockert, E.; Chen, Y.-T.; McGregor, D.; Jungbluth, A. Immunoreactivity for A103, an Antibody to Melan-A (Mart-1), in Adrenocortical and Other Steroid Tumors. Am. J. Surg. Pathol. 1998, 22, 57–63. [Google Scholar] [CrossRef] [PubMed]
- Ghorab, Z.; Jorda, M.; Ganjei, P.; Nadji, M. Melan A (A103) Is Expressed in Adrenocortical Neoplasms but Not in Renal Cell and Hepatocellular Carcinomas. Appl. Immunohistochem. Mol. Morphol. 2003, 11, 330–333. [Google Scholar] [CrossRef]
- Butterfield, L.H. Lessons learned from cancer vaccine trials and target antigen choice. Cancer Immunol. Immunother. 2016, 65, 805–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Habra, M.A.; Stephen, B.; Campbell, M.; Hess, K.; Tapia, C.; Xu, M.; Ahnert, J.R.; Jimenez, C.; Lee, J.E.; Perrier, N.D.; et al. Phase II clinical trial of pembrolizumab efficacy and safety in advanced adrenocortical carcinoma. J. Immunother. Cancer 2019, 7, 253. [Google Scholar] [CrossRef]
- Raj, N.P.; Zheng, Y.; Kelly, V.; Katz, S.; Chou, J.F.; Do, R.K.G.; Capanu, M.; Zamarin, D.; Ariyan, C.E.; Untch, B.R.; et al. Efficacy and safety of pembrolizumab in patients with advanced adrenocortical carcinoma. J. Clin. Oncol. 2019, 37, 4112. [Google Scholar] [CrossRef]
- Geoerger, B.; Kang, H.J.; Yalon-Oren, M.; Marshall, L.V.; Vezina, C.; Pappo, A.; Laetsch, T.W.; Petrilli, A.S.; Ebinger, M.; Toporski, J.; et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): Interim analysis of an open-label, single-arm, phase 1–2 trial. Lancet Oncol. 2020, 21, 121–133. [Google Scholar] [CrossRef]
- Head, L.; Kiseljak-Vassiliades, K.; Clark, T.J.; Somerset, H.; King, J.; Raeburn, C.; Albuja-Cruz, M.; Weyant, M.; Cleveland, J.; Wierman, M.; et al. Response to Immunotherapy in Combination with Mitotane in Patients with Metastatic Adrenocortical Cancer. J. Endocr. Soc. 2019, 3, 2295–2304. [Google Scholar] [CrossRef] [PubMed]
- Carneiro, B.A.; Konda, B.; Costa, R.B.; Costa, R.L.B.; Sagar, V.; Gursel, D.B.; Kirschner, L.S.; Chae, Y.K.; Abdulkadir, S.A.; Rademaker, A.; et al. Nivolumab in Metastatic Adrenocortical Carcinoma: Results of a Phase 2 Trial. J. Clin. Endocrinol. Metab. 2019, 104, 6193–6200. [Google Scholar] [CrossRef] [PubMed]
- Le Tourneau, C.; Hoimes, C.; Zarwan, C.; Wong, D.J.; Bauer, S.; Claus, R.; Wermke, M.; Hariharan, S.; Von Heydebreck, A.; Kasturi, V.; et al. Avelumab in patients with previously treated metastatic adrenocortical carcinoma: Phase 1b results from the JAVELIN solid tumor trial. J. Immunother. Cancer 2018, 6, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papewalis, C.; Fassnacht, M.; Willenberg, H.S.; Domberg, J.; Fenk, R.; Rohr, U.-P.; Schinner, S.; Bornstein, S.R.; Scherbaum, W.A.; Schott, M. Dendritic cells as potential adjuvant for immunotherapy in adrenocortical carcinoma. Clin. Endocrinol. 2006, 65, 215–222. [Google Scholar] [CrossRef]
- Liu-Chittenden, Y.; Jain, M.; Kumar, P.; Patel, D.; Aufforth, R.; Neychev, V.; Sadowski, S.; Gara, S.K.; Joshi, B.H.; Cottle-Delisle, C.; et al. Phase I trial of systemic intravenous infusion of interleukin-13-Pseudomonasexotoxin in patients with metastatic adrenocortical carcinoma. Cancer Med. 2015, 4, 1060–1068. [Google Scholar] [CrossRef] [Green Version]
- Ortmann, D.; Hausmann, J.; Beuschlein, F.; Schmenger, K.; Stahl, M.; Geissler, M.; Reincke, M. Steroidogenic Acute Regulatory (StAR)-Directed Immunotherapy Protects against Tumor Growth of StAR-Expressing Sp2-0 Cells in a Rodent Adrenocortical Carcinoma Model. Endocrinology 2004, 145, 1760–1766. [Google Scholar] [CrossRef] [Green Version]
- Majzner, R.G.; Theruvath, J.L.; Nellan, A.; Heitzeneder, S.; Cui, Y.; Mount, C.W.; Rietberg, S.P.; Linde, M.H.; Xu, P.; Rota, C.; et al. CAR T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors. Clin. Cancer Res. 2019, 25, 2560–2574. [Google Scholar] [CrossRef]
- Soysal, S.D.; Tzankov, A.; Muenst, S.E. Role of the Tumor Microenvironment in Breast Cancer. Pathobiology 2015, 82, 142–152. [Google Scholar] [CrossRef]
- Theocharis, S.; Tasoulas, J.; Masaoutis, C.; Kokkali, S.; Klijanienko, J. Salivary gland cancer in the era of immunotherapy: Can we exploit tumor microenvironment? Expert Opin. Ther. Targets 2020, 24, 1047–1059. [Google Scholar] [CrossRef] [PubMed]
- Yin, H.; Tang, Y.; Guo, Y.; Wen, S. Immune Microenvironment of Thyroid Cancer. J. Cancer 2020, 11, 4884–4896. [Google Scholar] [CrossRef] [PubMed]
Drug | Target | Number of Patients | Type of Study | Results | Reference |
---|---|---|---|---|---|
Pembrolizumab | PD-1 | 16 | Clinical Trial (Phase II) | PR in 12.5% of patients SD in 12.5% of patients | [101] |
39 | Clinical Trial (Phase II) | PR in 23% of patients SD in 17.9% of patients PSF = 2.1 mo OS = 24.9 | [102] | ||
4 (pediatric) | Clinical Trial (Phase II) | PR in two patients | [103] | ||
Pembrolizumab + Mitotane | PD-1 | 6 | Case Series | PR in two patients SD in four patients | [104] |
Nivolumab | PD-1 | 10 | Clinical Trial (Phase II) | PR in one patient SD in two patients PSF = 1.8 mo | [105] |
Avelumab | PD-L1 | 50 | Clinical Trial | PR in 6% of patients SD in 42% of patients PSF = 2.6 mo | [106] |
Nivolumab + Ipilimumab | PD-1/CTLA-4 | ongoing | Clinical Trial | ongoing |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Georgantzoglou, N.; Kokkali, S.; Tsourouflis, G.; Theocharis, S. Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success? Cancers 2021, 13, 1798. https://doi.org/10.3390/cancers13081798
Georgantzoglou N, Kokkali S, Tsourouflis G, Theocharis S. Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success? Cancers. 2021; 13(8):1798. https://doi.org/10.3390/cancers13081798
Chicago/Turabian StyleGeorgantzoglou, Natalia, Stefania Kokkali, Gerasimos Tsourouflis, and Stamatios Theocharis. 2021. "Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success?" Cancers 13, no. 8: 1798. https://doi.org/10.3390/cancers13081798
APA StyleGeorgantzoglou, N., Kokkali, S., Tsourouflis, G., & Theocharis, S. (2021). Tumor Microenvironment in Adrenocortical Carcinoma: Barrier to Immunotherapy Success? Cancers, 13(8), 1798. https://doi.org/10.3390/cancers13081798