Next Article in Journal
Exploring the In Vitro and In Vivo Therapeutic Potential of BRAF and MEK Inhibitor Combination in NRAS-Mutated Melanoma
Previous Article in Journal
Concordance between Three Homologous Recombination Deficiency (HRD) Assays in Patients with High-Grade Epithelial Ovarian Cancer
Previous Article in Special Issue
Advances in Molecular Pathophysiology and Targeted Therapy for Cushing’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Pituitary Tumors: Molecular Insights, Diagnosis, and Targeted Therapy

by
Kazunori Kageyama
1,* and
Mitsuru Nishiyama
2
1
Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Aomori 036-8562, Japan
2
Health Care Center, Kochi University, 1-5-2 Akebono-cho, Kochi 780-8520, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(23), 5526; https://doi.org/10.3390/cancers15235526
Submission received: 20 November 2023 / Accepted: 20 November 2023 / Published: 22 November 2023
(This article belongs to the Special Issue Pituitary Tumors: Molecular Insights, Diagnosis, and Targeted Therapy)
The anterior pituitary gland comprises a heterogeneous population of pituitary cells. Anterior cells are mainly characterized as corticotrophs, somatotrophs, lactotrophs, thyrotrophs, and gonadotrophs. Pituitary stem cells can differentiate into five hormone-producing cell types. This differentiation requires a complex cascade and is driven by specific transcription factors during development. Pituitary neuroendocrine tumors (PitNETs), formerly known as pituitary adenomas, are common intracranial tumors originating from the anterior pituitary neuroendocrine cells [1]. PitNETs are defined as autonomous/dysregulated cell proliferations or secretions of pituitary hormones. They present with various hormonal activities and clinical features, ranging from overt to subtle. In this Special Issue, we explore recent advances in molecular insights, diagnosis, and targeted therapy for PitNETs. The outlined papers include original clinical research articles and reviews on the pathophysiology, diagnosis, and potential treatment of PitNETs.
Oh et al. addressed the PitNET pathogenesis [2]. PitNETs are classified according to their lineage-restrictive pituitary transcription factors (TPIT for corticotroph tumors; PIT1 for somatotroph, lactotroph, and thyrotroph tumors; SF1 for gonadotroph tumors; the absence of PIT1, TPIT, and SF1 for null cell tumors). Less than 5% of PitNETs develop from germline mutations as a part of syndromic diseases or as a familial isolated pituitary adenoma [3], whereas the remaining 95% develop in the context of sporadic, somatic mutations in various genes that regulate the cell cycle, cell signaling, and transcriptional changes [4], lacking ubiquitous changes. Unlike hypersecreting PitNETs, nonfunctioning PitNETs may be found incidentally or with signs of mass effects rather than symptoms of excessive hormone secretion. Some nonfunctioning PitNETs are silent and show normal hormone secretion and anterior hormone or transcriptional factor-immunopositive staining without autonomous hormone secretion. Most silent PitNETs are silent gonadotrophs and all hypersecreting PitNET types have silent counterparts, such as silent lactotrophs, somatotrophs, and corticotroph tumors.
Tahara et al. reviewed pituitary incidentalomas [5]. The most common pituitary incidentalomas are PitNETs and Rathke cleft cysts. Surgical resection is recommended in cases of clinically nonfunctioning PitNETs with optic chiasm compression, whereas cystic lesions, such as Rathke cleft cysts, are followed up if the patients are asymptomatic. In contrast, certain pathological types of PitNETs, such as immature pluripotent PitNET lineage PIT1, Crooke’s cell tumor variant corticotroph PitNETs, and silent corticotroph PitNETs, are aggressive and therefore require careful monitoring.
Cushing’s disease is defined as the autonomous excess adrenocorticotropin (ACTH) secretion in corticotroph tumors (TPIT-lineage PitNETs) and excess cortisol production, with the clear manifestation of the clinical features of Cushing’s disease [6]. Mutations in the ubiquitin-specific protease (USP) 8 genes have been detected in Cushing’s disease [7,8,9]. This mutation increases enzyme activity, resulting in the excessive deubiquitination of epidermal growth factor receptor (EGFR) tyrosine kinase, disturbing its degradation [7]. EGFR expression levels positively correlate with ACTH production and cell proliferation in corticotrophic PitNETs. Phosphorylated EGFR expression has been reported in most corticotroph PitNETs [10], and an EGFR inhibitor could be effective in treating EGFR-related tumors. Gefitinib, a known EGFR tyrosine kinase inhibitor, has been found to suppress ACTH production and tumor growth in an experimental mouse model of Cushing’s disease using AtT-20 allografts [11]. Lapatinib, an active specific tyrosine kinase receptor inhibitor of both EGFR and p185her2/neu (HER2), significantly decreases ACTH production and corticotroph tumor cell proliferation [12].
A somatostatin analog with preferential affinity for somatostatin receptor type 5 (SSTR5) inhibits ACTH secretion in patients with ACTH-secreting PitNETs [13]. SSTR-mediated inhibition of phosphorylated extracellular signal-related kinases (ERK) via pasireotide decreases corticotrophic tumor cell proliferation [14]. Pasireotide may induce tumor shrinkage by directly acting on tumor cells [15]. Recently, researchers have found that SSTR5 is highly expressed in USP8-mutated tumors and pasireotide reportedly increases its antisecretory response in these tumors [16]. The overexpression of heat shock protein 90 (HSP90) has been reported in Cushing’s disease [17], which restrains the release of mature glucocorticoid receptors and causes partial glucocorticoid resistance. Silibinin, a C-terminal HSP90 inhibitor, restores glucocorticoid sensitivity [17]; therefore, an appropriate HSP90 inhibitor could be an effective treatment [18]. Some histone deacetylase inhibitors have been shown to block cell proliferation and ACTH synthesis in mouse AtT-20 and human corticotroph PitNETs [19,20,21,22,23]. The overexpression of the cell cycle regulator cyclin E and low expression of the cell cycle inhibitor tumor protein 27Kip1 (p27) have been observed in Cushing’s disease [24,25]. Cyclin E expression is correlated with p27 loss in human corticotroph PitNETs [26]. R-roscovitine, a pharmacological cyclin-dependent kinase 2 (CDK2)/cyclin E inhibitor, has also been shown to inhibit tumor growth in a mouse model of corticotroph tumors [27,28]. Overall, developments involving HSP90, histone deacetylase, and cyclin-dependent kinase inhibitors are expected for the treatment of Cushing’s disease [6].
In acromegaly, various genetic and epigenetic factors are involved in the somatotroph PitNET pathogenesis [29]. Somatic mutations of GNAS (Guanine nucleotide activating subunit) are the most prevalent cause of somatotroph PitNETs, whereas germline mutations in various genes, such as Aryl hydrocarbon receptor-interacting protein (AIP), Protein kinase cAMP-dependent type 1 regulatory subunit alpha (PRKAR1A), G protein-coupled receptor 101 (GPR101), GNAS, MEN1 (MENIN), Cyclin-dependent kinase inhibitor 1B (CDKN1B), Succinate dehydrogenase (SDHx), and MYC associated factor X (MAX), also cause somatotroph PitNETs. DNA hypomethylation and higher mRNA expression of potassium voltage-gated channel subfamily A regulatory beta subunit 2 (KCNAB2) have been observed in somatotrophs than in nonfunctioning pituitary PitNETs [30,31]. The pathophysiological relevance of miRNAs in somatotroph PitNETs has also been reported [29].
The entirety of the underlying mechanisms of somatotroph PitNETs has been clarified by multiple perspectives of the pan-genomic approach, including genome, transcriptome, methylome analyses, histological characterization, genomic instability, and possible miRNA involvement. Ferrés et al. determined the prognostic variables that facilitated predicting long-term postoperative outcomes to guide conceiving the most accurate acromegaly management practices [32]: younger age, higher preoperative growth hormone (GH), and insulin-like growth factor-1 levels, group 2b clinicopathological classification, Knosp’s grade IV, and magnetic resonance imaging. T2-weighted tumor hyperintensity and sparsely granulated cytokeratin expression patterns are associated with inferior postoperative outcomes in the long-term follow-up of patients with GH-secreting PitNETs.
Fukuhara et al. [33] updated the pathogenesis, diagnosis, and treatment of prolactinomas. Prolactinomas, comprising 30–50% of all PitNETs, are divided into two subgroups: familial and sporadic. Prolactinoma is the most frequent familial PitNET in patients with multiple endocrine neoplasia type 1 (MEN1) [34], whereas MEN1 gene mutations are also found in some sporadic prolactinomas. In addition, prolactinoma has been found in families with mutations in the PRKAR1A (Carney complex), CDKN1B (MEN4), or AIP (familial isolated pituitary adenoma: FIPA) genes [35]. Most prolactinomas can be treated using dopamine agonists. Temozolomide may be effective against malignant prolactinomas and a good response is obtained when O6-methylguanine-DNA methyltransferase immunostaining is negative. New medical treatments using estrogen receptor (ER) antagonists such as tamoxifen or raloxifene have also shown promising results [36] because ER is expressed in most surgical prolactinoma specimens. Recently, mammalian targets of rapamycin or tyrosine kinase inhibitors have been reported to be effective against aggressive prolactinomas [37,38].
Matsumoto et al. developed pituitary induction methods using human-induced pluripotent stem cells (hiPSCs), advancing the induction of pituitary glands from mouse embryonic stem cells [39,40]. Genome editing technologies using CRISPR/Cas systems have become standard methods. Additionally, the CRISPR/Cas system can introduce causative mutations from various malignant tumors into hPSCs. Therefore, combining hPSC-derived pituitary hormone-producing cells and genome-editing technologies may be useful for pituitary tumor research and developing novel drugs in the future.

Author Contributions

Conceptualization, K.K. and M.N.; writing—original draft preparation, K.K.; writing—review and editing, M.N. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Melmed, S. Pituitary-tumor endocrinopathies. N. Engl. J. Med. 2020, 382, 937–950. [Google Scholar] [CrossRef]
  2. Oh, J.Y.; Osorio, R.C.; Jung, J.; Carrete, L.; Choudhary, N.; Lad, M.; Saha, A.; Aghi, M.K. Transcriptomic profiles of normal pituitary cells and pituitary neuroendocrine tumor cells. Cancers 2022, 15, 110. [Google Scholar] [CrossRef]
  3. Spada, A.; Mantovani, G.; Lania, A.G.; Treppiedi, D.; Mangili, F.; Catalano, R.; Carosi, G.; Sala, E.; Peverelli, E. Pituitary tumors: Genetic and molecular factors underlying pathogenesis and clinical behavior. Neuroendocrinology 2022, 112, 15–33. [Google Scholar] [CrossRef]
  4. Melmed, S. Pathogenesis of pituitary tumors. Nat. Rev. Endocrinol. 2011, 7, 257–266. [Google Scholar] [CrossRef]
  5. Tahara, S.; Hattori, Y.; Suzuki, K.; Ishisaka, E.; Teramoto, S.; Morita, A. An overview of pituitary incidentalomas: Diagnosis, clinical features, and management. Cancers 2022, 14, 4324. [Google Scholar] [CrossRef]
  6. Takayasu, S.; Kageyama, K.; Daimon, M. Advances in molecular pathophysiology and targeted therapy for Cushing’s disease. Cancers 2023, 15, 496. [Google Scholar] [CrossRef]
  7. Reincke, M.; Sbiera, S.; Hayakawa, A.; Theodoropoulou, M.; Osswald, A.; Beuschlein, F.; Meitinger, T.; Mizuno-Yamasaki, E.; Kawaguchi, K.; Saeki, Y.; et al. Mutations in the deubiquitinase gene USP8 cause Cushing’s disease. Nat. Genet. 2015, 47, 31–38. [Google Scholar] [CrossRef]
  8. Perez-Rivas, L.G.; Theodoropoulou, M.; Ferraù, F.; Nusser, C.; Kawaguchi, K.; Stratakis, C.A.; Faucz, F.R.; Wildemberg, L.E.; Assié, G.; Beschorner, R.; et al. The gene of the ubiquitin-specific protease 8 is frequently mutated in adenomas causing Cushing’s disease. J. Clin. Endocrinol. Metab. 2015, 100, E997–E1004. [Google Scholar] [CrossRef]
  9. Ma, Z.Y.; Song, Z.J.; Chen, J.H.; Wang, Y.F.; Li, S.Q.; Zhou, L.F.; Mao, Y.; Li, Y.M.; Hu, R.G.; Zhang, Z.Y.; et al. Recurrent gain-of-function USP8 mutations in Cushing’s disease. Cell Res. 2015, 25, 306–317. [Google Scholar] [CrossRef]
  10. Theodoropoulou, M.; Arzberger, T.; Gruebler, Y.; Jaffrain-Rea, M.L.; Schlegel, J.; Schaaf, L.; Petrangeli, E.; Losa, M.; Stalla, G.K.; Pagotto, U. Expression of epidermal growth factor receptor in neoplastic pituitary cells: Evidence for a role in corticotropinoma cells. J. Endocrinol. 2004, 183, 385–394. [Google Scholar] [CrossRef]
  11. Fukuoka, H.; Cooper, O.; Ben-Shlomo, A.; Mamelak, A.; Ren, S.G.; Bruyette, D.; Melmed, S. EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. J. Clin. Investig. 2011, 121, 4712–4721. [Google Scholar] [CrossRef]
  12. Asari, Y.; Kageyama, K.; Sugiyama, A.; Kogawa, H.; Niioka, K.; Daimon, M. Lapatinib decreases the ACTH production and proliferation of corticotroph tumor cells. Endocr. J. 2019, 66, 515–522. [Google Scholar] [CrossRef]
  13. Colao, A.; Petersenn, S.; Newell-Price, J.; Findling, J.W.; Gu, F.; Maldonado, M.; Schoenherr, U.; Mills, D.; Salgado, L.R.; Biller, B.M.; et al. A 12-month phase 3 study of pasireotide in Cushing’s disease. N. Engl. J. Med. 2012, 366, 914–924. [Google Scholar] [CrossRef]
  14. Treppiedi, D.; Giardino, E.; Catalano, R.; Mangili, F.; Vercesi, P.; Sala, E.; Locatelli, M.; Arosio, M.; Spada, A.; Mantovani, G.; et al. Somatostatin analogs regulate tumor corticotrophs growth by reducing ERK1/2 activity. Mol. Cell. Endocrinol. 2019, 483, 31–38. [Google Scholar] [CrossRef]
  15. Theodoropoulou, M.; Stalla, G.K. Somatostatin receptors: From signaling to clinical practice. Front. Neuroendocrinol. 2013, 34, 228–252. [Google Scholar] [CrossRef]
  16. Albani, A.; Perez-Rivas, L.G.; Tang, S.; Simon, J.; Lucia, K.E.; Colón-Bolea, P.; Schopohl, J.; Roeber, S.; Buchfelder, M.; Rotermund, R.; et al. Improved pasireotide response in USP8 mutant corticotroph tumours in vitro. Endocr. Relat. Cancer 2022, 29, 503–511. [Google Scholar] [CrossRef]
  17. Riebold, M.; Kozany, C.; Freiburger, L.; Sattler, M.; Buchfelder, M.; Hausch, F.; Stalla, G.K.; Paez-Pereda, M.A.A. A C-terminal HSP90 inhibitor restores glucocorticoid sensitivity and relieves a mouse allograft model of Cushing disease. Nat. Med. 2015, 21, 276–280. [Google Scholar] [CrossRef]
  18. Sugiyama, A.; Kageyama, K.; Murasawa, S.; Ishigame, N.; Niioka, K.; Daimon, M. Inhibition of heat shock protein 90 decreases ACTH production and cell proliferation in AtT-20 cells. Pituitary 2015, 18, 542–553. [Google Scholar] [CrossRef]
  19. Nakada, Y.; Kageyama, K.; Sugiyama, A.; Desaki, R.; Takayasu, S.; Niioka, K.; Murasawa, S.; Ishigame, N.; Asari, Y.; Iwasaki, Y.; et al. Inhibitory effects of trichostatin A on adrenocorticotropic hormone production and proliferation of corticotroph tumor AtT-20 cells. Endocr. J. 2015, 62, 1083–1090. [Google Scholar] [CrossRef]
  20. Lu, J.; Chatain, G.P.; Bugarini, A.; Wang, X.; Maric, D.; Walbridge, S.; Zhuang, Z.; Chittiboina, P. Histone deacetylase inhibitor SAHA is a promising treatment of Cushing disease. J. Clin. Endocrinol. Metab. 2017, 102, 2825–2835. [Google Scholar] [CrossRef]
  21. Liu, S.; Cheng, H.; Kwan, W.; Lubieniecka, J.M.; Nielsen, T.O. Histone deacetylase inhibitors induce growth arrest, apoptosis, and differentiation in clear cell sarcoma models. Mol. Cancer Ther. 2008, 7, 1751–1761. [Google Scholar] [CrossRef]
  22. Hagiwara, R.; Kageyama, K.; Niioka, K.; Takayasu, S.; Tasso, M.; Daimon, M. Involvement of histone deacetylase 1/2 in adrenocorticotropic hormone synthesis and proliferation of corticotroph tumor AtT-20 cells. Peptides 2021, 136, 170441. [Google Scholar] [CrossRef] [PubMed]
  23. Hagiwara, R.; Kageyama, K.; Iwasaki, Y.; Niioka, K.; Daimon, M. Effects of tubastatin A on adrenocorticotropic hormone synthesis and proliferation of AtT-20 corticotroph tumor cells. Endocr. J. 2022, 69, 1053–1060. [Google Scholar] [CrossRef] [PubMed]
  24. Jordan, S.; Lidhar, K.; Korbonits, M.; Lowe, D.G.; Grossman, A.B. Cyclin D and cyclin E expression in normal and adenomatous pituitary. Eur. J. Endocrinol. 2000, 143, R1–R6. [Google Scholar] [CrossRef] [PubMed]
  25. Lidhar, K.; Korbonits, M.; Jordan, S.; Khalimova, Z.; Kaltsas, G.; Lu, X.; Clayton, R.N.; Jenkins, P.J.; Monson, J.P.; Besser, G.M.; et al. Low expression of the cell cycle inhibitor p27Kip1 in normal corticotroph cells, corticotroph tumors, and malignant pituitary tumors. J. Clin. Endocrinol. Metab. 1999, 84, 3823–3830. [Google Scholar] [CrossRef]
  26. Roussel-Gervais, A.; Bilodeau, S.; Vallette, S.; Berthelet, F.; Lacroix, A.; Figarella-Branger, D.; Brue, T.; Drouin, J. Cooperation between cyclin E and p27(Kip1) in pituitary tumorigenesis. Mol. Endocrinol. 2010, 24, 1835–1845. [Google Scholar] [CrossRef]
  27. Liu, N.A.; Jiang, H.; Ben-Shlomo, A.; Wawrowsky, K.; Fan, X.M.; Lin, S.; Melmed, S. Targeting zebrafish and murine pituitary corticotroph tumors with a cyclin-dependent kinase (CDK) inhibitor. Proc. Natl. Acad. Sci. USA 2011, 108, 8414–8419. [Google Scholar] [CrossRef]
  28. Liu, N.A.; Araki, T.; Cuevas-Ramos, D.; Hong, J.; Ben-Shlomo, A.; Tone, Y.; Tone, M.; Melmed, S. Cyclin E-mediated human proopiomelanocortin regulation as a therapeutic target for Cushing disease. J. Clin. Endocrinol. Metab. 2015, 100, 2557–2564. [Google Scholar] [CrossRef]
  29. Yamamoto, M.; Takahashi, Y. Genetic and epigenetic pathogenesis of acromegaly. Cancers 2022, 14, 3861. [Google Scholar] [CrossRef]
  30. Ling, C.; Pease, M.; Shi, L.; Punj, V.; Shiroishi, M.S.; Commins, D.; Weisenberger, D.J.; Wang, K.; Zada, G. A pilot genome-scale profiling of DNA methylation in sporadic pituitary macroadenomas: Association with tumor invasion and histopathological subtype. PLoS ONE 2014, 9, e96178. [Google Scholar] [CrossRef]
  31. Ashton, C.; Rhie, S.K.; Carmichael, J.D.; Zada, G. Role of KCNAB2 expression in modulating hormone secretion in somatotroph pituitary adenoma. J. Neurosurg. 2020, 134, 787–793. [Google Scholar] [CrossRef] [PubMed]
  32. Ferrés, A.; Reyes, L.; Di Somma, A.; Topczewski, T.; Mosteiro, A.; Guizzardi, G.; De Rosa, A.; Halperin, I.; Hanzu, F.; Mora, M.; et al. The prognostic-based approach in growth hormone-secreting pituitary neuroendocrine tumors (PitNET): Tertiary reference center, single senior surgeon, and long-term follow-up. Cancers 2022, 15, 267. [Google Scholar] [CrossRef] [PubMed]
  33. Fukuhara, N.; Nishiyama, M.; Iwasaki, Y. Update in pathogenesis, diagnosis, and therapy of prolactinoma. Cancers 2022, 14, 3604. [Google Scholar] [CrossRef] [PubMed]
  34. Pieterman, C.R.C.; Valk, G.D. Update on the clinical management of multiple endocrine neoplasia type 1. Clin. Endocrinol. 2022, 97, 409–423. [Google Scholar] [CrossRef]
  35. Schernthaner-Reiter, M.H.; Trivellin, G.; Stratakis, C.A. MEN1, MEN4, and Carney complex: Pathology and molecular genetics. Neuroendocrinology 2016, 103, 18–31. [Google Scholar] [CrossRef]
  36. Choudhary, C.; Hamrahian, A.H.; Bena, J.F.; Recinos, P.; Kennedy, L.; Dobri, G. The effect of raloxifene on serum prolactin level in patients with prolactinoma. Endocr. Pract. 2019, 25, 684–688. [Google Scholar] [CrossRef]
  37. Cooper, O.; Bonert, V.S.; Rudnick, J.; Pressman, B.D.; Lo, J.; Salvatori, R.; Yuen, K.C.J.; Fleseriu, M.; Melmed, S. EGFR/ErbB2-targeting lapatinib therapy for aggressive prolactinomas. J. Clin. Endocrinol. Metab. 2021, 106, e917–e925. [Google Scholar] [CrossRef]
  38. Zhang, D.; Way, J.S.; Zhang, X.; Sergey, M.; Bergsneider, M.; Wang, M.B.; Yong, W.H.; Heaney, A.P. Effect of everolimus in treatment of aggressive prolactin-secreting pituitary adenomas. J. Clin. Endocrinol. Metab. 2019, 104, 1929–1936. [Google Scholar] [CrossRef]
  39. Matsumoto, R.; Suga, H.; Arima, H.; Yamamoto, T. Disease modeling of pituitary adenoma using human pluripotent stem cells. Cancers 2022, 14, 3660. [Google Scholar] [CrossRef]
  40. Suga, H.; Kadoshima, T.; Minaguchi, M.; Ohgushi, M.; Soen, M.; Nakano, T.; Takata, N.; Wataya, T.; Muguruma, K.; Miyoshi, H.; et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature 2011, 480, 57–62. [Google Scholar] [CrossRef]
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

Kageyama, K.; Nishiyama, M. Pituitary Tumors: Molecular Insights, Diagnosis, and Targeted Therapy. Cancers 2023, 15, 5526. https://doi.org/10.3390/cancers15235526

AMA Style

Kageyama K, Nishiyama M. Pituitary Tumors: Molecular Insights, Diagnosis, and Targeted Therapy. Cancers. 2023; 15(23):5526. https://doi.org/10.3390/cancers15235526

Chicago/Turabian Style

Kageyama, Kazunori, and Mitsuru Nishiyama. 2023. "Pituitary Tumors: Molecular Insights, Diagnosis, and Targeted Therapy" Cancers 15, no. 23: 5526. https://doi.org/10.3390/cancers15235526

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