Browse
Instituto de Endocrinología IEMYR, Quito, Ecuador
Maastricht University, Maastricht, The Netherlands
Search for other papers by Jaime Guevara-Aguirre in
Google Scholar
PubMed
Search for other papers by Gabriela Peña in
Google Scholar
PubMed
Search for other papers by William Acosta in
Google Scholar
PubMed
Search for other papers by Gabriel Pazmiño in
Google Scholar
PubMed
Search for other papers by Jannette Saavedra in
Google Scholar
PubMed
Search for other papers by Lina Soto in
Google Scholar
PubMed
Search for other papers by Daniela Lescano in
Google Scholar
PubMed
Search for other papers by Alexandra Guevara in
Google Scholar
PubMed
Search for other papers by Antonio W D Gavilanes in
Google Scholar
PubMed
The relationship between growth hormone (GH) excess and cancer is a controversial matter. Until 2016, most studies in patients with acromegaly found links with colon and thyroid neoplasms. However, recent studies found increased risks in gastric, breast, and urinary tract cancer also. Concordantly, clinical situations where GH and insulin-like growth facto-I deficits exist are indeed associated with diminished malignancy incidence. In line with these observations, gain-of-function mutations of various enzymes belonging to the GH and IGF-I signaling pathways have been associated with increased carcinogenesis; similarly, loss-of-function mutations of other enzymes that usually work as tumor repressors are also associated with augmented cancer risk. In a study performed in Ecuador, it was demonstrated that subjects in the Ecuadorian cohort with Laron syndrome (ELS), who have a mutant GH receptor and greatly diminished GH and IGF-I signaling, display diminished incidence of cancer. Along with absent action of GH and IGF-I, ELS individuals also have low serum insulin levels and decreased insulin resistance. Furthermore, hyperglycemia and hyperinsulinemia are indispensable for fast cell mitosis, including that of those cells present in the benign and malignant neoplasms. Notably, and despite their obesity, subjects with the ELS display normoglycemia and hypo-insulinemia, along with diminished incidence of malignancies. We believe that the dual low-IGF-I/low insulin serum levels are responsible for the cancer protection, especially considering that the insulin/INSR signaling is a central site for energy generation in the form of ATP and GDP, which are indispensable for all and every GH/IGF-I physiologic as well as pathologic events.
Division of Pulmonary and Critical Care Medicine, Asante Health System, Medford, Oregon, USA
Search for other papers by Bilal F Samhouri in
Google Scholar
PubMed
Search for other papers by Thorvardur R Halfdanarson in
Google Scholar
PubMed
Search for other papers by Chi Wan Koo in
Google Scholar
PubMed
Department of Respiratory Medicine, St Vincent’s University Hospital, Elm Park, Dublin, Ireland
Search for other papers by Cormac McCarthy in
Google Scholar
PubMed
Search for other papers by Eunhee S Yi in
Google Scholar
PubMed
Search for other papers by Charles F Thomas in
Google Scholar
PubMed
Search for other papers by Jay H Ryu in
Google Scholar
PubMed
Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia (DIPNECH) is a rare, but increasingly recognized entity that primarily affects middle-aged and elderly women. It is characterized by abnormal proliferation of pulmonary neuroendocrine cells (PNECs) and is considered a preinvasive lesion for carcinoid tumorlets/tumors. Sometimes, DIPNECH is accompanied by constrictive bronchiolitis which usually manifests as chronic cough and/or dyspnea, along with airflow limitation on spirometry. The telltale imaging sign of DIPNECH is the presence of multiple noncalcified pulmonary nodules and mosaic attenuation on CT. However, these clinico-radiologic features of DIPNECH are characteristic but nonspecific; thus, histopathologic confirmation is usually necessary. DIPNECH has an indolent course and only rarely leads to respiratory failure or death; progression to overt neuroendocrine tumor (carcinoid) of the lung occurs in a minority of patients. Of available therapies, somatostatin analogs and mechanistic target of rapamycin inhibitors are the most promising. In this review, we provide an update regarding the diagnosis and management of DIPNECH and describe critical gaps in our understanding of this entity, including the central terms ‘diffuse’ and ‘idiopathic.’ We also summarize the inconsistencies in definitions employed by recent studies and discuss the pitfalls of the DIPNECH definitions proposed by the World Health Organization in 2021. In this context, we propose an objective and reproducible radio-pathologic case definition intended for implementation in the research realm and seeks to enhance homogeneity across cohorts. Furthermore, we discuss aspects of PNECs biology which suggest that PNEC hyperplasia may contribute to the pathogenesis of phenotypes of lung disease aside from constrictive bronchiolitis and carcinoid tumorlets/tumors. Finally, we steer attention to some of the most pressing and impactful research questions awaiting to be unraveled.
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Andreas Venizelos in
Google Scholar
PubMed
Department of Clinical Science, University of Bergen, Bergen, Norway
Search for other papers by Halfdan Sorbye in
Google Scholar
PubMed
Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
Search for other papers by Hege Elvebakken in
Google Scholar
PubMed
Search for other papers by Aurel Perren in
Google Scholar
PubMed
Search for other papers by Inger Marie B Lothe in
Google Scholar
PubMed
Search for other papers by Anne Couvelard in
Google Scholar
PubMed
Search for other papers by Geir Olav Hjortland in
Google Scholar
PubMed
Department of Medical Radiation Physics, Lund University, Lund, Sweden
Search for other papers by Anna Sundlöv in
Google Scholar
PubMed
Search for other papers by Johanna Svensson in
Google Scholar
PubMed
Search for other papers by Harrish Garresori in
Google Scholar
PubMed
Search for other papers by Christian Kersten in
Google Scholar
PubMed
Department of Oncology, St. Olavs Hospital, Trondheim, Norway
Search for other papers by Eva Hofsli in
Google Scholar
PubMed
Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
Search for other papers by Sönke Detlefsen in
Google Scholar
PubMed
Search for other papers by Lene W Vestermark in
Google Scholar
PubMed
Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
Search for other papers by Morten Ladekarl in
Google Scholar
PubMed
Search for other papers by Elizaveta Mitkina Tabaksblat in
Google Scholar
PubMed
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Stian Knappskog in
Google Scholar
PubMed
High-grade gastroenteropancreatic (HG-GEP) neuroendocrine neoplasms (NENs) are highly aggressive cancers. The molecular etiology of these tumors remains unclear, and the prevalence of pathogenic germline variants in patients with HG-GEP NENs is unknown. We assessed sequencing data of 360 cancer genes in normal tissue from 240 patients with HG-GEP NENs; 198 patients with neuroendocrine carcinomas (NECs) and 42 with grade 3 neuroendocrine tumors (NET G3). Applying strict criteria, we identified pathogenic germline variants and compared the frequency with previously reported data from 33 different cancer types. We found a recurrent MYOC variant in three patients and a recurrent MUTYH variant in two patients, indicating that these genes may be important underlying risk factors for HG-GEP NENs when mutated. Further, germline variants were found in canonical tumor-suppressor genes, such as TP53, RB1, BRIP1 and BAP1. Overall, we found that 4.5% of patients with NEC and 9.5% of patients with NET G3 carry germline pathogenic or highly likely pathogenic variants. Applying identical criteria for variant classification in silico to mined data from 33 other cancer types, the median percentage of patients carrying pathogenic or highly likely pathogenic variants was 3.4% (range: 0–17%). The patients with NEC and pathogenic germline variants had a median overall survival of 9 months, similar to what is generally expected for metastatic GEP NECs. A patient with NET G3 and pathogenic MUTYH variant had much shorter overall survival than expected. The fraction of HG-GEP NENs with germline pathogenic variants is relatively high, but still <10%, meaning that that germline mutations cannot be the major underlying cause of HG-GEP NENs.
Search for other papers by Christopher A Galifi in
Google Scholar
PubMed
Search for other papers by Teresa L Wood in
Google Scholar
PubMed
Despite decades of research presenting insulin-like growth factor-1 receptor (IGF1R) as an attractive target for cancer therapy, IGF1R inhibitors ultimately failed in clinical trials. This was surprising due to the known cancer-promoting functions of IGF1R, including stimulation of cell invasion, proliferation, and survival. Discourse in the literature has acknowledged that a lack of patient stratification may have impacted the success of IGF1R-inhibitor trials. This argument alludes to the possibility that IGF1R function may be contingent on tumor type and cellular composition. Looking into the known roles of IGF1R, it becomes clear that this receptor interacts with a multitude of different proteins and even has tumor-suppressing functions. IGF1R is implicated in both cell–cell and cell–surface adhesion dynamics, and the effects of either IGF1R downregulation or pharmacological inhibition on cellular adhesion remain poorly understood. In turn, adhesion receptors modulate IGF1R signaling. In addition, our understanding of IGF1R function in tumor-associated immune and stromal cells is lacking, which could contribute to the overwhelming failure of IGF1R inhibitors in the clinic. In this review, we re-investigate clinical trial data to make connections between the failure of these drugs in human cancer patients and the understudied facets of IGF1R function. We describe lesser-known and potentially tumor-suppressive functions of IGF1R that include promoting cell–cell adhesion through E-cadherin, augmenting a pro-inflammatory macrophage phenotype, and stimulating B cells to produce immunoglobulins. We also highlight the important role of adhesion receptors in regulating IGF1R function, and we use this information to infer stratification criteria for selecting patients that might benefit from IGF1R inhibitors.
European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, Florence, Italy
Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, Florence, Italy
Istituto Nazionale Biostrutture e Biosistemi (I.N.B.B.), via delle Medaglie D’Oro, Rome, Italy
Search for other papers by Giulia Cantini in
Google Scholar
PubMed
Search for other papers by Elena Niccolai in
Google Scholar
PubMed
European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, Florence, Italy
Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, Florence, Italy
Search for other papers by Letizia Canu in
Google Scholar
PubMed
Search for other papers by Leandro Di Gloria in
Google Scholar
PubMed
Search for other papers by Simone Baldi in
Google Scholar
PubMed
European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, Florence, Italy
Search for other papers by Arianna Pia Propato in
Google Scholar
PubMed
European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, Florence, Italy
Search for other papers by Laura Fei in
Google Scholar
PubMed
Search for other papers by Giulia Nannini in
Google Scholar
PubMed
Search for other papers by Soraya Puglisi in
Google Scholar
PubMed
Search for other papers by Gabriella Nesi in
Google Scholar
PubMed
Search for other papers by Matteo Ramazzotti in
Google Scholar
PubMed
Search for other papers by Amedeo Amedei in
Google Scholar
PubMed
European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, Florence, Italy
Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, Florence, Italy
Istituto Nazionale Biostrutture e Biosistemi (I.N.B.B.), via delle Medaglie D’Oro, Rome, Italy
Search for other papers by Michaela Luconi in
Google Scholar
PubMed
The infiltrating microbiota represents a novel cellular component of the solid tumour microenvironment that can influence tumour progression and response to therapy. Adrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy for which mitotane (MTT) treatment represents the first-line therapy, though its efficacy is limited to a therapeutic window level (14–20 mg/L). Novel markers able to predict those patients who would benefit from MTT therapy are urgently needed to improve patient’s management. The aim of our study was to evaluate the presence of intratumoural bacterial microbiota DNA in 26 human ACC tissues vs 9 healthy adrenals; moreover, the association between the relative bacterial composition profile, the tumour mass characteristics and MTT ability to reach high circulating levels in the early phase of treatment, were explored. We found the presence of bacterial DNA in all adrenal samples from both tumours and healthy cortex specimens, documenting significant differences in the microbial composition between malignancy and normal adrenals: in detail, the ACC tissues were characterised by a higher abundance of the Proteobacteria phylum (especially the Pseudomonas and Serratia genera). In addition, the Proteobacteria’s low abundance was negatively associated with tumour size, Ki67 and cortisol secretion. MTT levels reached higher levels at 9 months in ACC patients with high abundance of Proteobacteria, Pseudomonas and Serratia and with low abundance of Bacteroidota, Firmicutes and Streptococcus. These findings are the first indication that human ACCs are characterised by infiltrating bacteria and their specific abundance profile seems to influence the increase in circulating MTT levels at 9 months.
Search for other papers by Jared S Rosenblum in
Google Scholar
PubMed
Search for other papers by Herui Wang in
Google Scholar
PubMed
Search for other papers by Matthew A Nazari in
Google Scholar
PubMed
Search for other papers by Zhengping Zhuang in
Google Scholar
PubMed
Search for other papers by Karel Pacak in
Google Scholar
PubMed
This article is a summary of the plenary lecture presented by Jared Rosenblum that was awarded the Manger Prize at the Sixth International Symposium on Pheochromocytoma/Paraganglioma held on 19–22 October 2022 in Prague, Czech Republic. Herein, we review our initial identification of a new syndrome of multiple paragangliomas, somatostatinomas, and polycythemia caused by early postzygotic mosaic mutations in EPAS1, encoding hypoxia-inducible factor 2 alpha (HIF-2α), and our continued exploration of new disease phenotypes in this syndrome, including vascular malformations and neural tube defects. Continued recruitment and close monitoring of patients with this syndrome as well as the generation and study of a corresponding disease mouse model as afforded by the pheochromocytoma/paraganglioma translational program at the National Institutes of Health has provided new insights into the natural history of these developmental anomalies and the pathophysiologic role of HIF-2α. Further, these studies have highlighted the importance of the timing of genetic defects in the development of related disease phenotypes. The recent discovery and continued study of this syndrome has not only rapidly evolved our understanding of pheochromocytoma and paraganglioma but also deepened our understanding of other developmental tumor syndromes, heritable syndromes, and sporadic diseases.
Search for other papers by Xuan Chen in
Google Scholar
PubMed
Search for other papers by Sixuan Liu in
Google Scholar
PubMed
Search for other papers by Xue Peng in
Google Scholar
PubMed
Search for other papers by Xiangyun Zong in
Google Scholar
PubMed
Anti-Müllerian hormone (AMH) is produced and secreted by granulosa cells of growing follicles, and its main role is to inhibit the recruitment of primordial follicles, reduce the sensitivity of follicles to follicle-stimulating hormone (FSH), and regulate FSH-dependent preantral follicle growth. It has become an effective indicator of ovarian reserve in clinical practice. Research on AMH and its receptors in recent years has led to a better understanding of its role in breast cancer. AMH specifically binds to anti-Müllerian hormone receptor II (AMHRII) to activate downstream pathways and regulate gene transcription. Since AMHRII is expressed in breast cancer cells and triggers apoptosis, AMH/AMHRII may play an important role in the occurrence, treatment, and prognosis of breast cancer, which needs further research. The AMH level is a potent predictor of ovarian function after chemotherapy in premenopausal breast cancer patients older than 35 years, either for ovarian function injury or ovarian function recovery. Moreover, AMHRII has the potential to be a new marker for the molecular typing of breast cancer and a new target for breast cancer treatment, which may be a link in the downstream pathway after TP53 mutation.
Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Jiajun Wu in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Juyong Liang in
Google Scholar
PubMed
Search for other papers by Ruiqi Liu in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Tian Lv in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Kangyin Fu in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Liehao Jiang in
Google Scholar
PubMed
Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Wenli Ma in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Yan Pan in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Zhuo Tan in
Google Scholar
PubMed
Search for other papers by Qing Liu in
Google Scholar
PubMed
Search for other papers by Weihua Qiu in
Google Scholar
PubMed
Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Search for other papers by Minghua Ge in
Google Scholar
PubMed
Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
Department of Thyroid and Breast Surgery, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
Search for other papers by Jiafeng Wang in
Google Scholar
PubMed
Anlotinib-mediated angiogenic remodeling was delineated in various tumors. Meanwhile, we previously showed that anlotinib inhibited tumor angiogenesis in anaplastic thyroid cancer (ATC). However, the potential role of anlotinib on cell lethality in ATC remains an enigma. Herein, we found that anlotinib inhibited the viability, proliferation, and migration of KHM-5M, C643, and 8505C cells in a dose-dependently manner. Under anlotinib treatment, PANoptosis (pyroptosis, apoptosis, and necroptosis) markers were not changed; however, ferroptosis targets (transferrin, HO-1, FTH1, FTL, and GPX4) were significantly downregulated. ROS levels also increased in a concentration-dependent manner after anlotinib treatment in KHM-5M, C643, and 8505C cells. In addition, protective autophagy was activated in response to anlotinib, and autophagic blockade potentiated anlotinib-mediated ferroptosis and antitumor effects in vitro and in vivo. Our new discovery identified autophagy-ferroptosis signaling pathway which provides mechanistic insight into anlotinib-mediated cell death, and synergistic combination therapy may help develop new ATC treatment strategies.
Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
Search for other papers by Elizabeth J de Koster in
Google Scholar
PubMed
Search for other papers by Willem E Corver in
Google Scholar
PubMed
Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
Search for other papers by Lioe-Fee de Geus-Oei in
Google Scholar
PubMed
Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, the Netherlands
Department of Biomedical Sciences and Humanitas Clinical and Research Centre, Department of Nuclear Medicine, Humanitas University, Milan, Italy
Search for other papers by Wim J G Oyen in
Google Scholar
PubMed
Search for other papers by Dina Ruano in
Google Scholar
PubMed
Search for other papers by Abbey Schepers in
Google Scholar
PubMed
Search for other papers by Marieke Snel in
Google Scholar
PubMed
Search for other papers by Tom van Wezel in
Google Scholar
PubMed
Search for other papers by Dennis Vriens in
Google Scholar
PubMed
Search for other papers by Hans Morreau in
Google Scholar
PubMed
Whole chromosome instability with near-whole genome haploidization (GH) and subsequent endoreduplication is considered a main genomic driver in the tumorigenesis of oncocytic cell thyroid neoplasms (OCN). These copy number alterations (CNA) occur less frequently in oncocytic thyroid adenoma (OA) than in oncocytic carcinoma (OCA), suggesting a continuous process. The current study described the CNA patterns in a cohort of 30 benign and malignant OCN, observed using a next-generation sequencing (NGS) panel that assesses genome-wide loss of heterozygosity (LOH) and chromosomal imbalances using 1500 single-nucleotide polymorphisms (SNPs) across all autosomes and the X chromosome in DNA derived from cytological and histological samples. Observed CNA patterns were verified using multiparameter DNA flow cytometry with or without whole-genome SNP array analysis and lesser-allele intensity-ratio (LAIR) analysis. On CNA–LOH analysis using the NGS panel, GH-type CNA were observed in 4 of 11 (36%) OA and in 14 of 16 OCA (88%). Endoreduplication was suspected in 8 of 16 (50%) OCA, all with more extensive GH-type CNA (P < 0.001). Reciprocal chromosomal imbalance type CNA, characterized by (imbalanced) chromosomal copy number gains and associated with benign disease, were observed in 6 of 11 (55%) OA and one equivocal case of OCA. CNA patterns were different between the histopathological subgroups (P < 0.001). By applying the structured interpretation and considerations provided by the current study, CNA–LOH analysis using an NGS panel that is feasible for daily practice may be of great added value to the widespread application of molecular diagnostics in the diagnosis and risk stratification of OCN.
Search for other papers by Zvi Laron in
Google Scholar
PubMed
Search for other papers by Haim Werner in
Google Scholar
PubMed
Many clinical and experimental studies have implicated the growth hormone (GH)–insulin-like growth factor (IGF-1) axis with the progression of cancer. The epidemiological finding that patients with Laron syndrome (LS), the best-characterized disease under the spectrum of congenital IGF-1 deficiencies, do not develop cancer is of major scientific and translational relevance. The evasion of LS patients from cancer emphasizes the central role of the GH–IGF-1 system in cancer biology. To identify genes that are differentially expressed in LS and that might provide a biological foundation for cancer protection, we have recently conducted genome-wide profiling of LS patients and normal controls. Analyses were performed on immortalized lymphoblastoid cell lines derived from individual patients. Bioinformatic analyses identified a series of genes that are either over- or under-represented in LS. Differential expression was demonstrated in a number of gene families, including cell cycle, metabolic control, cytokine–cytokine receptor interaction, Jak-STAT and PI3K-AKT signaling, etc. Major differences between LS and controls were also noticed in pathways associated with cell cycle distribution, apoptosis, and autophagy. The identification of novel downstream targets of the GH–IGF-1 network highlights the biological complexity of this hormonal system and sheds light on previously unrecognized mechanistic aspects associated with GH–IGF-1 action in the cancer cell.