Targeting growth hormone in cancer: future perspectives

in Endocrine-Related Cancer
Authors:
Yue Wang Liggins Institute, University of Auckland, Auckland, New Zealand
Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand

Search for other papers by Yue Wang in
Current site
Google Scholar
PubMed
Close
,
Stephen M F Jamieson Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand

Search for other papers by Stephen M F Jamieson in
Current site
Google Scholar
PubMed
Close
, and
Jo K Perry Liggins Institute, University of Auckland, Auckland, New Zealand
Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand

Search for other papers by Jo K Perry in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-4418-947X

Correspondence should be addressed to J K Perry: j.perry@auckland.ac.nz

This paper is part of a themed collection on the role of Growth Hormone in Endocrine Cancers. The Guest Editor for this collection was John Kopchick, Ohio University, USA.

Free access

Sign up for journal news

Decades of published research support a role for growth hormone (GH) in cancer. Accordingly, there is increasing interest in targeting GH in oncology, with GH antagonists exhibiting efficacy in xenograft studies as single agents and in combination with anticancer therapy or radiation. Here we discuss challenges associated with using growth hormone receptor (GHR) antagonists in preclinical models and considerations for translation, such as the identification of predictive biomarkers for selecting patients and for monitoring drug efficacy. Ongoing research will determine whether suppressing GH signalling pharmacologically will also reduce the risk of developing cancer. An increase in GH-targeted drugs in preclinical development will ultimately provide new tools to test anticancer efficacy of blocking the GH signalling pathway.

Abstract

Decades of published research support a role for growth hormone (GH) in cancer. Accordingly, there is increasing interest in targeting GH in oncology, with GH antagonists exhibiting efficacy in xenograft studies as single agents and in combination with anticancer therapy or radiation. Here we discuss challenges associated with using growth hormone receptor (GHR) antagonists in preclinical models and considerations for translation, such as the identification of predictive biomarkers for selecting patients and for monitoring drug efficacy. Ongoing research will determine whether suppressing GH signalling pharmacologically will also reduce the risk of developing cancer. An increase in GH-targeted drugs in preclinical development will ultimately provide new tools to test anticancer efficacy of blocking the GH signalling pathway.

Introduction

Observations in humans and animals with elevated or disrupted growth hormone (GH) signalling clearly support a role for GH in cancer development (Guevara-Aguirre et al. 2020, Werner et al. 2020, Duran-Ortiz et al. 2021). Clinically, increased GH levels have been identified in the sera of cancer patients and in numerous tumour types, with GH and growth hormone receptor (GHR) expression levels correlating with poorer clinical and histopathological parameters (Emerman et al. 1985, Mazzoccoli et al. 1999, Wu et al. 2011, Kong et al. 2016, Perry et al. 2017, Arumugam et al. 2019, Kaseb et al. 2022). There is also extensive preclinical evidence implicating GH signalling in multiple aspects of cancer progression (Chhabra et al. 2011, Brittain et al. 2017, Perry et al. 2017, Chesnokova & Melmed 2019, Kopchick et al. 2022) and treatment resistance (Basu & Kopchick 2019, Cheng et al. 2021). Such observations highlight the potential of this pathway as a therapeutic target. However, this has not yet translated into clinical trials. Encouragingly though, two studies have reported anticancer efficacy for a GHR antagonist in individual patients with liver cancer and male breast cancer in a patient with acromegaly (Leporati et al. 2015, Kaseb et al. 2022).

Systemic release of GH from the anterior pituitary regulates hepatic release of insulin-like growth factor-1 (IGF1), which like GH has been implicated in cancer (Cao & Yee 2021) (Fig. 1). GH also has autocrine and paracrine effects within the tumour (Perry et al. 2006, 2017, Harvey et al. 2015). Local GH expression is observed in cancer cells and cells within the tumour microenvironment, with autocrine GH having demonstrably more potent oncogenic effects on cancer cells compared to exogenous GH treatment (Kaulsay et al. 1999, Zhu et al. 2005, Perry et al. 2006). GH actions are initiated by interaction with a cell surface GHR homodimer, which is a class 1 cytokine receptor, through two asymmetric binding sites on GH. Binding causes a rotational change in the GHR transmembrane domain, facilitating phosphorylation of two JAK2 molecules that are associated with the cytoplasmic domain of the receptor (Brooks et al. 2014, Waters & Brooks 2015). In addition to activating the cell surface GHR, GH also activates the related prolactin receptor (PRLR) at the cell membrane (Bartke & Kopchick 2015) and crosstalks with other hormone and growth factor receptors, resulting in a complex multi-component signal transduction network (Liu et al. 2016, Lu et al. 2019, Frank 2020). Therapeutic strategies targeting GH will therefore need to consider receptor crosstalk and systemic and local functions.

Figure 1
Figure 1

Regulation of GH and therapeutic blockade. GH and IGF1 affect tumour growth through endocrine, autocrine and paracrine mechanisms. GH is secreted from the anterior pituitary with release regulated by the hypothalamic hormones, growth hormone releasing hormone (GHRH) and somatostatin (SST), and ghrelin, which is predominantly produced by the stomach. Increased circulating GH promotes release of IGF1 from the liver and other target tissues. Additional regulation is provided by a feedback loop mechanism (+ and −), with high circulating GH and IGF1 inhibiting GH secretion. GH and IGF1 can also be expressed in extra-pituitary tissues and tumours, where they have local autocrine/paracrine actions on GHR- and IGF1R-expressing tumour cells, as well as cells in the tumour microenvironment. Inhibition of GH actions can be achieved through direct targeting of the GHR and by inhibition of GHR protein production. GH actions can also be blocked indirectly by inhibitors/agonists of hormones that regulate GH release from the pituitary. *GHRH, antagonists, ghrelin antagonists, and SST analogues also directly target tumour cells expressing their respective receptors. GHBP, growth hormone-binding protein; GHR, growth hormone receptor; GHRH, growth hormone releasing hormone; IGF1, insulin-like growth factor 1; SST, somatostatin. Created with BioRender.com.

Citation: Endocrine-Related Cancer 30, 9; 10.1530/ERC-23-0033

The many reported mechanisms by which GH promotes cancer (promoting cell proliferation and survival, epithelial to mesenchymal transition, cell migration and invasion, cell senescence, tumour growth, angiogenesis and metastasis, and drug and radiation resistance) have been reviewed previously (Chhabra et al. 2011, Brittain et al. 2017, Perry et al. 2017, Basu & Kopchick 2019, Chesnokova & Melmed 2019, Kopchick et al. 2022) and elsewhere in this special issue. The purpose of this review is to discuss recent advances and some of the considerations when translating GH inhibition into the clinical oncology setting.

Clinical studies

Recent years have seen a rapid increase in the number of clinical studies supporting the role for GH in cancer, with higher GH and GHR levels tending to be associated with poorer clinical outcome, although this varies by cancer. Good clinical correlations are observed for cancers such as breast, endometrial, melanoma and hepatocellular carcinoma (HCC) (Wu et al. 2011, Sustarsic et al. 2013, Kong et al. 2016, Perry et al. 2017, Kaseb et al. 2022) (Tables 1 and 2). In HCC, increased GH is associated with poorer outcome, whereas the converse is seen for GHR (as described later on). However, study designs and the methodology used to assess GH and GHR levels can vary greatly, making it difficult to compare across studies.

Table 1

Clinical assessment of GH and GHR levels in mammary carcinoma.

Breast cancer
References Clinical outcome Study methodology
(Emerman et al. 1985) Among the serum of 42 patients with breast cancer, 40% had increased GH and 17% had elevated PRL; total lactogens/(GH+PRL) ratio was greater in patients with breast cancer compared to controls a Radioimmunoassay; bioassay
(Love et al. 1991) Increasing age was associated with decreased GH. No differences in prolactin or GH levels among premenopausal women with breast cancer (18), healthy women with a strong family history of breast cancer (n = 23) and healthy women (n = 39) Radioimmunoassay; bioassay
(Mol et al. 1995) GH was expressed in normal and neoplastic mammary gland tissue RT-PCR; IHC
(Mertani et al. 1998) Co-expression of GHR and PRLR in all breast cancer samples, with similar expression levels ISH; IHC
(Gebre-Medhin et al. 2001) GHR expression was increased in patients with breast cancer a RT-PCR; WB; IHC
(Raccurt et al. 2002) GH expression was increased in metastatic mammary carcinoma with de novo stromal expression in proliferative disorders of the mammary gland a ISH; RT-PCR
(Menashe et al. 2010) GH-induced intracellular signalling pathway was the third most highly associated with breast cancer susceptibility among 421 pathways containing 3962 genesa GWAS/pathway analysis
(Wu et al. 2011) Tumoral expression of GH mRNA was significantly associated with lymph node metastasis, tumour stage, epidermal growth factor receptor 2 and proliferative index in breast cancer. GH and PRL expression correlated with worse relapse-free survival and overall survival in patients with breast cancer a ISH; IHC; Kaplan–Meier and Cox regression
(Chiesa et al. 2011) GH mRNA expression was present in 52% of clinically derived primary mammary cancer cell lines and GHR mRNA was expressed in all primary mammary cancer cell lines. In EpCAM-positive breast cancer, GH mRNA was not significantly associated with cancer grade, stage, lymph node metastasis or patient age RT-PCR; immunofluorescence
(Lombardi et al. 2014) GHR was expressed in the normal mammary epithelium. Expansion of a GHR-positive cell population in 72% of ductal carcinoma in situ lesions. There was a positive correlation between GHR and ER expressiona Immunofluorescence;

tissue microarrays
(Leporati et al. 2015) Case report: a 72-year-old male with acromegaly and breast cancer had normalisation of IGF1 with pegvisomant therapy. The patient stopped pegvisomant and started on anastrozole due to metastases 4 years after breast cancer surgery, but the metastatic lesion increased, and the patient was shifted to tamoxifen and restarted on pegvisomant. The size of the metastatic lesion reduced, and a further reduction was observed in the subsequent 24-month follow-up CT scan
(Hamann et al. 2019) GH concentration was significantly lower in early breast cancer N/A
(Arumugam et al. 2019) IHC demonstrated increased GHR expression was associated with lower ER expression (n = 72). In ER tumours (n = 671), high GHR mRNA was associated with poorer survival a IHC; KM plotter; Oncomine
(Dabrosin & Dabrosin 2020) Extracellular GH and IGFBPs higher in ER+ breast cancer than normal adjacent breast tissue (n = 10) a Multiplex PEA
(Zhu et al. 2020) GHR mRNA was highly expressed in breast tumour (n = 12) compared to adjacent normal tissues (n = 12)a qPCR

aIndicates studies that show an increase in GHR or GH expression in tumour samples or serum; upregulation of GHR signalling in tumour samples; or a positive association of GH or GHR with poorer prognosis.

AR V7, androgen receptor splice variant 7; CT scan, computerised tomography scan; EpCAM, epithelial cell adhesion molecule; ER, oestrogen receptor; GEM, gemcitabine; GWAS, genome-wide association study; Hep C, hepatitis C; HCC, hepatocellular carcinoma; IHC, immunohistochemistry; IGF1, insulin-like growth factor-1; IGFBP, IGF-binding protein; ISH, in situ hybridisation; KM, Kaplan–Meier; mAb, monoclonal antibody; MRI, magnetic resonance imaging; PEG, pegvisomant; PRL, prolactin; PRLR, prolactin receptor; PEA, proximity extension assay; qPCR, quantitative real-time PCR; STAT5, signal transducer and activator of transcription 5; TCGA, The Cancer Genome Atlas; WB, western blot.

Table 2

Clinical assessment of GH and GHR levels in hepatocellular carcinoma.

Hepatocellular carcinoma (HCC)
References Clinical outcome Study methodology
(Chang et al. 1990) GHR not detected in HCC or cirrhotic tissue, all control liver tissues had high-affinity and low-affinity GHR Radioreceptor assays
(Liu et al. 2003) The receptor-binding capacity of the GHR in HCC was lower than in normal liver, and it was negatively associated with tumour size and disease stage Radioreceptor assays
(García-Caballero et al. 2000) The expression of GHR and PRLR was increased in HCC samples compared to normal liver a ISH; IHC
(Kong et al. 2016) Both GH and PRL were increased in HCC compared with non-neoplastic liver tissues on gene and protein levels. GH expression significantly associated with tumour size and grade. Expression of GH and PRL was associated with worse relapse-free and overall survival in HCC patientsa ISH; PCR; IHC
(Lin et al. 2021) 200 patients with chronic Hep C and HCC; GHR mRNA was significantly lower in Hep C virus-related HCC qPCR
(Mai et al. 2021) GHRmRNA was downregulated in HCC patients MustSeq
(Cai et al. 2022b ) 11 differentially expressed ageing-related genes (including GHR) correlated with HCC patient prognosis TCGA and GEO databases
(Basu et al. 2022) GHR and IGF1 mRNA levels were strongly correlated in HCC patients (n = 371). IGF1R was correlated with poorer survival, and GH-regulated ABCB1 and IGF1-regulated ABCC1 were negatively correlated with survival for HCC Bioinformatic analyses of TCGA datasets
(Kaseb et al. 2022) High plasma GH associated with poor clinical outcome. Two HCC patients with high plasma GH had stabilised disease after treatment with pegvisomant combined with sorafenib following progression on sorafenib alone a ELISA; MRI
(Abu El-Makarem et al. 2022) Cirrhotic patients with HCC had considerably lower hepatic expression of the GHR, STAT5 and IGF1 proteins compared to cirrhotic patients without HCC and healthy controls IHC
(Mohamed et al. 2022) HCC patients in GH-low group (n = 18) had significantly superior 1-year survival rate and overall survival compared to patients in GH-high group (n = 19) a ELISA; Kaplan–Meier survival analysis
(Kodama et al. 2022) Significant inverse association between GHR gene expression with patient survival qPCR and RNA-Seq
(Guo et al. 2022) High GHR mRNA expression in tumours from the low-risk group Gene expression, TCGA
(Zhang et al. 2022) An eight-gene signature including GHR was used in predicting the clinical outcome of HCC patients Transcriptome analysis
The GHR gene was downregulated in high-risk group KM analysis; qPCR
(He et al. 2022) HCC patients with high expression of the GHRmRNA had longer survival time Gene expression

aIndicates studies that show an increase in GHR or GH expression in tumour samples or serum; upregulation of GHR signalling in tumour samples; or a positive association of GH or GHR with poorer prognosis.

CT scan, computerised tomography scan; EpCAM, epithelial cell adhesion molecule; ER, oestrogen receptor; GWAS, genome-wide association study; Hep C, hepatitis C; HCC, hepatocellular carcinoma; IHC, immunohistochemistry; IGF1, insulin-like growth factor-1; IGFBP, IGF-binding protein; ISH, in situ hybridisation; KM, Kaplan–Meier; MRI, magnetic resonance imaging; PRL, prolactin; PRLR, prolactin receptor; PEA, proximity extension assay; qPCR, quantitative real-time PCR; STAT5, signal transducer and activator of transcription 5; TCGA, The Cancer Genome Atlas.

Because of the endocrine, autocrine and paracrine functions of GH, measurements of GH mRNA or protein levels can be made in tumour tissues and circulating GH in serum or plasma (Perry et al. 2013, 2017). Gene expression profiling of GH and/or GHR from large clinical cohorts is available to researchers through online databases such as The Cancer Genome Atlas, but the data lacks protein expression and circulating GH levels. In addition, while low levels of GH expression can have a profound effect on cancer cell phenotype, low mRNA levels are not easily detected using methods such as RNA sequencing and require other techniques such as in situ hybridisation. Furthermore, GH is a secreted protein, so measurements of protein expression in tissues can be problematic but can be achieved using immunohistochemistry. In the circulation, GH is released in pulses from the pituitary so measurements in serum or plasma may be inaccurate depending on the time the sample is taken. Despite this, correlations between serum GH and outcome have been observed in HCC, breast, gastric, lung and colon cancer and this approach may warrant further investigation (Emerman et al. 1985, Mazzoccoli et al. 1999, Triantafillidis et al. 2003, Kaseb et al. 2022).

Test specificity is also an issue. The GH gene locus is composed of five genes that code for evolutionarily related hormones (GH(GH1), placental GH (GH2) and the placental lactogens (CSH1, CSH2, CSHL1)) that share a high degree of sequence similarity at the DNA and amino acid level (Liao et al. 2018). Differentiating between these five genes can be difficult when using quantitative PCR, and antibodies to GH often cross-react with placental GH and placental lactogen. GH-related hormones may also be deregulated in cancer cells so ELISAs and antibodies used in immunohistochemistry and western blotting should be rigorously validated to ensure specificity.

The increasing number of publications in this area is encouraging but there is still an acute need for large-scale clinical studies profiling GH and GHR expression, circulating GH, clinicopathological features and survival outcome in patients to inform clinical trials.

Cancer variants

GH and GHR are encoded respectively by the GH1 and GHR genes. Perhaps surprisingly given the described role for GH in cancer, cancer-associated driver mutations in the GH1 and GHR gene are not common. Notably, the GHR-P495T mutation is the only GHR variant for which a role in cancer promotion is supported by functional data (Chhabra et al. 2018). The single nucleotide polymorphisms (SNP) contributing to GHR-P495T (rs6183) was initially identified from a lung cancer genome wide association study (GWAS) (minor allele frequency = 0.001, odds ratio = 12.98, P = 0.0019) (Rudd et al. 2006). Cells harbouring the GHR-P495T mutation have prolonged GHR signalling due to reduced SOCS2 binding at amino acid position 495 which leads to impaired cellular degradation and turnover of the mutant receptor (Chhabra et al. 2018). Cancer-associated variants have been identified in genes that regulate GH expression and release, such as the somatostatin gene promoter (Wagner et al. 2006). A small number of cancer variants linked to GH/IGF1 genes have also been identified. For example, SNPs in the highly polymorphic proximal promoter of the GH1 gene have been linked to breast cancer risk (Wagner et al. 2007), while a SNP in intron 4 of GH1 was associated with lower circulating GH and IGF1, and reduced risk of colorectal cancer (Hasegawa et al. 2000, le Marchand et al. 2002, Millar et al. 2010, Shi et al. 2014).

However, caution needs to be taken when assigning gene targets to non-coding SNPs. Although a disease-associated SNP may be in close proximity to a gene of interest (for example in the promoter, intron or 3’-UTR), proximity should not be mistaken for association. Intragenic SNPs residing in regulatory regions such as enhancers can affect the expression of other neighbouring genes, or even genes located much further away on the same or different chromosomes (Sanyal et al. 2012, Pudjihartono et al. 2022). An example of this is a breast cancer-associated SNP, rs6171, in the GH1 promoter. The GH locus control region and promoter act as a complex regulatory hub that coordinates expression of the five genes within the GH locus through chromosome looping (Tsai et al. 2016) but may also regulate the expression of numerous other genes proximal and distal to the locus (Jain et al. 2020). Indeed, the rs6171 SNP is routinely linked to GH due to its location in the promoter and is associated with changes in expression of the GH locus gene, CSH2. But it is also associated with changes in expression of several genes flanking the locus (CD79B, DDX42, FTSJ3 and TCAM1P) (Jain et al. 2020, supplementary data; GTEx), two of which (FTSJ3 and TCAM1P) have been linked to cancer (Manning et al. 2020, Zhu et al. 2022). Thus, GH1 SNPs such as rs6171 may impact on cancer risk through other genes in addition to GH1 or besides.

Expression changes in individual genes or effect sizes of single SNPs may be small in isolation, but when combined these may have more impact on cancer risk. Body mass index, exercise and ethnicity may modify the association between some GH-related SNPs and cancer risk (le Marchand et al. 2002, Khoury-Shakour et al. 2008, Shi et al. 2014, Gao et al. 2015). This highlights the importance of pathway analyses rather than focussing on single SNPs. For example, GWAS are useful for identifying common-low penetrance risk alleles but tend to identify relatively few highly significant loci. But combining pathway analysis with existing GWAS data can discover new candidate genes. Such an approach identified the GH intracellular signalling pathway as one of three pathways (out of 421 investigated) that were highly associated with breast cancer susceptibility (Menashe et al. 2010).

Confirming functionality for non-coding SNPs is a complex process but necessary in order to avoid mis-assigning SNPs to a proximal gene. Finally, it should be noted that these types of analyses also have the potential to identify new non-coding SNPs that can act independently or in combination to change GH1 transcript levels, thus potentiating GH effects in cancer cells.

Challenges using GHR antagonists in animal models

Decades of published research strongly support a role for GH in cancer; however, only a few preclinical oncology studies involving pharmacological inhibition of GHR have been carried out in animals. This can be attributed to a lack of access to suitable long-acting antagonists. Of the studies that have been performed, results have been largely promising, with tumour growth inhibition observed with monotherapy GHR antagonist treatment and in combination with chemotherapy or radiation. Tumour models include breast, colorectal, meningeal, prostate, endometrial gastric, pancreatic and hepatocellular cancer (McCutcheon et al. 2001, Dagnaes-Hansen et al. 2004, Divisova et al. 2006, Lombardi et al. 2014, Evans et al. 2016, Brody et al. 2021, Kaseb et al. 2022, Meng et al. 2022, Unterberger et al. 2022) (Table 3 and reviewed in this issue).

Table 3

Pharmacological inhibition of GH signalling in xenograft studies.

Cancer type Cell line Dose Outcome References
Meningioma Primary meningiomas (n = 15) (↓) Pegvisomant, 45 mg/kg, daily Serum IGF1: 19% reduction (McCutcheon et al. 2001)
Monotherapy inhibition of tumour growth
Colon COLO 205 (↓) Pegvisomant, 60 mg/kg, every second day Serum IGF1: 56–64% reduction (Dagnaes-Hansen et al. 2004)
HT-29 (-) Reduced COLO 205 volume with monotherapy, not HT-29
Breast MCF-7 (↓) Pegvisomant, 100 or 250 mg/kg, daily Serum IGF1: 75% reduction (injection daily), 62% reduction (every other day), 54% (every 3 days) (Divisova et al. 2006)
MDA-MD-231 (-) Reduced MCF-7 tumour volume at 100 & 250 mg/kg monotherapy; no effect on MDA-MB-231 or MDA-MB-435
MDA-MB-435 (-)
Breast Patient-derived breast cancer (↓) Pegvisomant, 40 mg/kg, daily, for 2 weeks IGF-1: not described (Lombardi et al. 2014)
Monotherapy with pegvisomant-inhibited tumour growth
Endometrial RL95-2 (↓) Pegvisomant, 100 mg/kg, every second day Serum IGF1: 45–76% reduction (Evans et al. 2016)
Radiation + PEG significantly delayed tumour growth; no monotherapy effect
Prostate 22Rv1 (-) Pegvisomant, 100 mg/kg, 3 times weekly IGF1: liver and tumour expression reduced (WB) (Recouvreux et al. 2017)
No effect on tumour growth with monotherapy or combination with enzalutamide; decreased expression of AR V7
Prostate Pten-mutant mouse prostate cancer cell (↓) Pegvisomant, 100 mg/kg, 3 times per week IGF1: 18% reduction (3 days after the final injection) (Unterberger et al. 2022)
Monotherapy-reduced Pten-mutant prostate tumour growth
Hepatocellular HepG2 (↓) Pegvisomant, 100 mg/kg, daily IGF1: ~80% reduction (Kaseb et al. 2022)
Monotherapy-reduced tumour growth; reduction was more pronounced when combined with sorafenib
Gastric SUN-16 (↓) Pegvisomant, 250 mg/kg, daily IGF-1: not described (Meng et al. 2022)
Monotherapy: inhibition of tumour growth
Pancreatic PANC1 (↓) Pegvisomant, 10 mg/kg/day; Compound G, 10 mg/kg/day IGF1: not described (Brody et al. 2021)
Monotherapy: PEG and compound G decreased tumour growth
GEM combined with PEG or compound G was more efficacious
Breast MCF-7 (↓) GHR/PRLR neutralising mAb; 15 or 30 mg/kg, twice weekly IGF1: not described (Chen et al. 2020)
T-47D (↓) Monotherapy: inhibited the growth of T-47D and MCF-7
Breast MDA-MB-231 (↓) Small-molecule BM 001–003, 1 and 5 mg/kg, 3 times per week IGF1: 73% reduction (van der Velden et al. 2022)
Monotherapy: all three reduced tumour growth; BM001 was most effective

(↓) indicates cell lines for which efficacy with GHR antagonism was observed either as a single agent or in combination therapy, while (–) indicates no effect.

AR V7, androgen receptor splice variant 7; GEM, gemcitabine; IGF1, insulin-like growth factor-1; mAb, monoclonal antibody; PEG, pegvisomant; PRLR, prolactin receptor; WB, western blot.

A major obstacle to preclinical studies in animal models which also limits access to suitable antagonists is the species specificity of the drug. Pegvisomant is the only clinically available GHR antagonist. It is approved by the FDA for the treatment of acromegaly, a disease associated with elevated circulating GH and IGF1 (Neggers et al. 2016). Pegvisomant is a PEGylated derivative of B2036, a mutant GH protein containing a G120K mutation at binding site 2 which abrogates activation of the GHR, as well as eight additional mutations in binding site 1 (Kopchick et al. 2002, 2014, van der Lely & Kopchick 2006). Both GH and B2036 have a very short circulating half-life (15–20 min). To improve half-life, B2036 is conjugated with 5 kDa polyethylene glycol (PEG) via amine residues resulting in a heterogenous mix of conjugates with 4–6 PEG moieties. PEGylation reduces renal filtration and extends the serum half-life of pegvisomant in humans to 72 h, but it comes at the cost of a significantly lower binding affinity for the mouse Ghr (Kopchick et al. 2002, 2014, van der Lely & Kopchick 2006). Consequently, very high doses of pegvisomant are required to achieve effective Ghr inhibition and reduce circulating IGF1 in rodents. For example, the dose used in humans to normalise IGF1 levels is 10–30 mg daily subcutaneous injections (~0.1–0.4 mg/kg for an average person) (Jen et al. 2013). However, the equivalent dose used in rodents is several hundred-fold higher with doses of 45–250 mg/kg/day (Table 3). Furthermore, as for many biotherapeutics, the circulating half-life is shorter in rodents vs humans so more frequent dosing is required (Mordenti et al. 1991).

Cancer studies that have used pegvisomant are summarised in Table 3. Combined, with the challenges that receptor species specificity poses to xenograft studies, there is a clearly a need for the development of GHR antagonists that have better efficacy against the mouse Ghr. Arguably, by using antagonists that have poor efficacy in rodents and as a consequence reduced anticancer efficacy, potentially effective therapies may be discontinued before they even have a chance of being evaluated in humans.

Another approach to investigate mechanisms of GHR antagonism in cancer progression is to use genetic models as an in vivo preclinical platform (Swanson & Unterman 2002, Kopchick et al. 2014, Qian et al. 2020, Lantvit et al. 2021, Basu et al. 2022, Haque et al. 2022). For example, the Kopchick lab used Ghr knockout mice or mice transgenic for a Ghr antagonist to assess the efficiency of combining chemotherapy with Ghr antagonism in mouse melanoma and HCC. Their results indicated that Ghr antagonism sensitised tumours to anticancer therapy (e.g. cisplatin and sorafenib) by downregulation of ATP-binding cassette multidrug efflux transporters (Qian et al. 2020, Basu et al. 2022). Swanson and Unterman found that GH-deficient spontaneous dwarf rats were resistant to the development of carcinogen-induced mammary tumours (Swanson & Unterman 2002). Another recent study incorporated mice with global or liver-specific disruption of the Ghr gene and induced HCC using diethylnitrosamine. Only 5.6% of mice with global Ghr deletion (Ghr−/−) developed tumours, compared with 93.5% of wild-type mice. Mice with liver-specific Ghr disruption also had significantly less tumours in the liver (Haque et al. 2022). While results from genetic studies are persuasive, deficiency in GH signalling, for example, through Gh1/Ghr gene disruption or constitutive expression of a GHR/Ghr antagonist, would be expected to affect initial tumour establishment and growth prior to treatment when compared with wild-type animals which are used as a control. A comparison with a treatment setting in patients is therefore limited. Alternatively, inducible deletion of the Ghr gene or expression of an antagonist in transgenic animals can be used. However, leaky expression of the transgene is common in inducible models which is likely to still affect tumour establishment compared to wild-type animals. These types of studies are still extremely useful, particularly in the absence of accessible long-acting inhibitors and clearly support a role for GH in tumour development and progression. But pharmacological inhibition is more likely to yield relevant data on translation, particularly if the drug effectively targets both mouse and human GH signalling.

Another consideration for preclinical studies is the mode of GH action. GH has endocrine, autocrine and paracrine effects (Fig. 1), with both systemic and tumour-derived GH accelerating cancer progression (Harvey et al. 2015, Perry et al. 2017, Lu et al. 2019). In addition, systemic release of GH from the anterior pituitary increases circulating IGF1 which is also implicated in many malignancies, particularly breast cancer (Cao & Yee 2021). Inhibiting GHR signalling would therefore be expected to have additional benefits via suppressing systemic IGF1. Clinical trials with agents that target the IGF1 receptor (IGF1R) have not been encouraging but this is attributed to multiple reasons including interaction with insulin receptor signalling and recent evidence that the role of the IGF1R in tumour development and phenotype may be context-dependent (Cao & Yee 2021, Bulatowicz & Wood 2022).

Taking endocrine, autocrine and paracrine actions into account when investigating GHR antagonism is therefore important, particularly in the context of rodent xenograft models where tumours are derived from human cancer cell lines and cells of murine origin in the tumour microenvironment. Due to species specificity, mouse Gh does not activate the human GHR (Souza et al. 1995). However, systemic Gh is the key regulator of Igf1 release from the liver into the circulation. Mouse Igf1 exhibits cross-species activity so can affect human cancer cells expressing human IGF1R and mouse cells expressing Igfr1 in the tumour microenvironment (Perry et al. 2013). But the endocrine system is not the only secretion pathway for GH. GH is also secreted locally from cancer and microenvironment cells, such as endothelial, immune and fibroblast cells, and affects tumour progression through autocrine/paracrine effects on neighbouring cells (Chesnokova & Melmed 2019, Lu et al. 2019).

Studies from in vivo xenograft experiments across different cancer models demonstrate that GHR antagonism can inhibit the growth of certain tumour types (Table 3). It is likely that this is due to the combined effects of supressing endocrine Gh/Igf1 and autocrine/paracrine effects of GH/Gh/IGF1/Igf1. This is harder to achieve in xenograft models due to the species specificity of GH and the antagonists (as described above) but is an important consideration as it more closely mimics the human clinical setting. Indeed, for preclinical models to effectively predict clinical activity, therapies will need to block GHR and Ghr to a similar degree. However, from a mechanistic point, the contribution that systemic vs autocrine/paracrine GH secretion plays in tumour progression is certainly of interest. Species-specific inhibitors will help dissect out the relative contributions of systemic and autocrine GH secretion in xenograft experiments.

The need for new drugs: current strategies targeting GH signalling and secretion

Strategies targeting GHR signalling

While pegvisomant is an extremely effective human GHR inhibitor, there has been no indication that the drug is being repurposed for oncology applications. The only cancer-related clinical study reported was a phase 1 dose-escalation trial conducted in 2007 to assess the clinical efficacy of pegvisomant in individuals with normal GH/IGF1 levels to guide dosing regimen for further evaluation in cancer patients. In this trial pegvisomant was well tolerated at high doses over 14 days (Yin et al. 2007). Access to pegvisomant for research purposes is also limited which has hampered the types of in vivo preclinical studies that can be conducted. The lack of access is partially driving a growing interest among researchers and clinicians in the development of novel inhibitors. Encouragingly, there has been a lot of recent activity in preclinical drug development, which should eventually expand the options available for clinical and research applications (Lu et al. 2019). We anticipate this will intensify, particularly given the interest in GHR signalling as a target for extending healthy lifespan (Longo et al. 2015, van der Spoel et al. 2016, Duran-Ortiz et al. 2021).

Generation of long-acting biological therapeutics such as pegvisomant has been the preferred and most effective approach to ensure selectivity (pegvisomant has no off-target effects against the related PRLR (Goffin et al. 1999)). Recently, variants of pegvisomant have been reported with alteration in the amino acid sequence and/or PEGylation at specific sites on the protein with larger PEG moieties (Brody et al. 2020, Wang et al. 2020, 2021). These include compound G, which is site-specifically PEGylated at 2 sites on the protein core (Brody et al. 2020, 2021). Compound G and pegvisomant exhibited single-agent efficacy against pancreatic cancer xenografts and reduced tumour growth in combination with gemcitabine, compared with monotherapy (Brody et al. 2021). Ongoing development of PEGylated protein GHR antagonists will likely utilise site-specific PEGylation as this yields a more homogenous drug during manufacture compared with the amine-conjugated approach used for pegvisomant. Another approach for generating long-acting therapeutics is by making large fusion proteins. For example, fusion of a GHR antagonist to a variant of GH-binding protein (GHBP) generated a potent antagonist with extended serum half-life (Wilkinson et al. 2016).

Another inhibitor that is under development is the antisense oligonucleotide, ATL1103 (Antisense Therapeutics). ATL1103 significantly reduced average serum IGF1 levels in patients with acromegaly in a phase II clinical trial (Trainer et al. 2018). Additional approaches include neutralising anti-GHR antibodies (Jiang et al. 2011, Siebel et al. 2014, Sun et al. 2015, Chen et al. 2020). An antibody reported recently by Chen et al. is a dual-function antagonist targeting both the GHR and PRLR that blocks signalling from both receptors and attenuates T-47D and MCF-7 tumour growth in vivo (Chen et al. 2020). Peptide antagonists are also an attractive approach due to more convenient manufacturing processes. A novel 16 amino acid helical peptide antagonist of the GHR (S1H) was recently reported that inhibits GH-dependent STAT5 phosphorylation in cell lines (Basu et al. 2021). A second bicyclic 16 amino acid peptide GHR antagonist (AZP-3813, Amolyt Pharma) is being developed as a potential treatment for acromegaly for use in combination with somatostatin analogues.

The GHR has not been considered a particularly good druggable target for small-molecule development. Targeting protein–protein interactions (PPIs), such as the GH–GHR interface, is difficult, as the contact surfaces tend to be large, flat and featureless which can negatively impact on the binding affinity of small molecules and their selectivity for the target. Despite challenges, recent high-profile successes targeting PPIs (Nero et al. 2014, Lu et al. 2020) and small-molecule inhibitors of the structurally related PRLR have been published which bind near the prolactin receptor PPIs (Borcherding et al. 2021). Thus, the likelihood of identifying GHR-inhibiting small molecules is more encouraging. An earlier report of a small-molecule GHR inhibitor most likely acts downstream of the receptor (Rosengren et al. 2007). But another approach by van der Velden et al. identified molecules that may inhibit GHR protein synthesis in the endoplasmic reticulum (van der Velden et al. 2022). This may be a better approach for cancer therapy as it avoids potential challenges when targeting autocrine GH; it has been suggested that receptor antagonists such as pegvisomant may not be as effective at inhibiting autocrine GH, as autocrine GH interacts with the GHR and initiates signalling in the endoplasmic reticulum and Golgi before it reaches the cell surface (van den Eijnden & Strous 2007, Strous et al. 2020). Given earlier studies from Peter Lobie’s group demonstrating that autocrine GH promotes cancer progression more effectively than exogenous GH treatment (Kaulsay et al. 1999, Mukhina et al. 2004, Zhu et al. 2005), effective targeting of autocrine GH will be an important clinical consideration. However, inhibiting intracellular autocrine GH alone may not be sufficient. B2036/pegvisomant can inhibit autocrine GH that is secreted into the media by breast cancer cells (Kaulsay et al. 2001). In our hands, stimulation with exogenous GH still activates GHR signalling in cell lines expressing endogenous autocrine GH (unpublished). Similarly, primary human mammary carcinoma cells expressing endogenous GH proliferate in response to exogenous GH treatment, and this can be blocked with a GHR antagonist (Chiesa et al. 2011). It is therefore feasible that both GH-induced signalling via cell surface receptor activation and intracellular GHR activation are necessary for full autocrine action by GH (Chhabra et al. 2011). Certainly, clarification regarding the mechanism by which autocrine GH has a greater role in cancer promotion than endocrine/systemic GH will be important to inform which inhibition strategies are more appropriate for oncology applications.

In addition to human antagonists, a mouse Ghr antagonist should be considered for studies in rodents. Given the promising results obtained with pegvisomant in tumour studies, it is likely that a mouse Ghr antagonist will be more effective due to more potent GHR inhibition/IGF1 suppression. Alternately, animal models humanised for the GH and GHR genes can be considered. A humanised GH1 mouse has been reported previously (Ariyasu et al. 2019). As observed with the human GH signalling system, human GH in humanised mice will activate the mouse Prlr (Bartke & Kopchick 2015) and therefore provides a means of investigating combinations of GHR and PRLR inhibitors in cancer.

Strategies targeting GH secretion

Strategies that regulate endocrine GH secretion may also be beneficial in cancer therapy. Hormones that control GH release from the anterior pituitary include hypothalamic hormones, GH-releasing hormone (stimulatory; GHRH) and somatostatin (inhibitory), and the predominantly gastric hormone, ghrelin (stimulatory) (Fig. 1). GH secretion can be inhibited by somatostatin analogues and dopamine agonists that are used clinically, as well as GHRH and ghrelin antagonists that are in development (Costantini et al. 2011, Melmed et al. 2018, Lu et al. 2019).

Somatostatin analogues were developed for acromegaly and are recommended as the first-line medical treatment for patients with uncontrolled disease following surgery or for those who do not undergo surgery (Melmed et al. 2018). Since then, these drugs have demonstrated benefit in the clinical management of neuroendocrine tumours that overexpress somatostatin receptors (SSTRs) and may also have application in other types of cancer (Rai et al. 2015, Kawasaki et al. 2023). Somatostatin inhibits hormone and growth factor secretion, apoptosis, cell proliferation and angiogenesis. The receptors for somatostatin are a family of five G protein-coupled receptors (GPCRs). First-generation somatostatin analogues, octreotide and lanreotide, are standard-of-care options for patients with advanced unresectable neuroendocrine tumours (Kawasaki et al. 2023). Anticancer efficacy is mediated by direct interaction with SSTRs on tumour cells, with potential additional mechanisms through reduced systemic GH/IGF1 and secretion of other hormones (Wolin 2012). Octreotide and lanreotide have higher affinity for SSTR2 than the other four receptors and inhibit GH secretion and, to a lesser extent, thyroid-stimulating hormone, adrenocorticotrophin and prolactin secretion (Labadzhyan & Melmed 2022).

GHRH and its receptor are also expressed across many different cancer types (Schally et al. 2008). For example, high levels of the GHRH receptor are associated with reduced 10-year overall survival rate in patients with locally advanced rectal cancer (Fodor et al. 2023). Several generations of well-characterised GHRH antagonists have been described that exhibit anticancer activity in vitro and in preclinical tumour models (Schally & Varga 2006, Schally et al. 2008, Zarandi et al. 2017, Cai et al. 2022a ). As with somatostatin analogues, mechanisms of action include direct inhibition of autocrine/paracrine effects on tumours, as well as indirect actions through reduced endocrine release of GH/IGF1 (Schally et al. 2008).

Another key hormone that controls GH secretion is ghrelin which regulates energy homeostasis and is a potent stimulator of GH release. Ghrelin acts through interaction with GH secretagogue receptor (GHSR), a GPCR which is expressed in the hypothalamus and pituitary (Yanagi et al. 2018). Small-molecule and peptide GHSR antagonists have been described. However, some exhibit unexpected behaviour with both antagonistic and agonist properties (Price et al. 2021). For example, GSK1614343 and BIM-28131 inhibit GH secretion in vivo but stimulate other ghrelin functions such as food intake (Costantini et al. 2011, Hassouna et al. 2013). GHSR signalling is very complex; there are many ligands, and receptor activity and pathway activation can be modified by heterodimerisation and crosstalk with numerous other transmembrane proteins (Price et al. 2021, Gross et al. 2023). As with GHRH and somatostatin, GHSR and its ligands are associated with tumour development and progression in multiple tumour types. Whether ghrelin antagonists will be beneficial as anticancer agents is still under investigation (Soleyman-Jahi et al. 2019, Kotta et al. 2022).

Looking forward: translational considerations

While there is now considerable evidence that GHR signalling plays an important role in cancer, these observations have not yet translated into clinical trials of GHR antagonists in patients. However, there have been anecdotal reports of tumour stabilisation and/or regression in individual cases treated with pegvisomant: two HCC patients and a male breast cancer patient with acromegaly (Leporati et al. 2015, Kaseb et al. 2022).

There are important factors to consider as the field turns towards the translation of GHR antagonists into clinical trials, particularly considering 94% of oncology drugs tested in clinical trials never obtain FDA approval (Haslam et al. 2023). Leading causes for failure in clinical trials include a lack of clinical efficacy, toxicities, trial design and a lack of biomarkers (Wong et al. 2019, Sun et al. 2022). In terms of clinical efficacy and toxicity, using pegvisomant or derivative drugs may be an advantage as pegvisomant has already been demonstrated to be a highly potent antagonist in the clinical setting that is selective for GHR and is well tolerated in normal individuals when given at high doses for short periods (Goffin et al. 1999, Yin et al. 2007). Efficacy studies in cancer clinical trials will be of interest to evaluate not only if pegvisomant has anticancer efficacy, but also whether its low toxicity profile is maintained over the longer treatment periods that might be needed in an oncology setting.

Crucially important is also the study design and selection of the tumour type and treatment modality, Monotherapy efficacy has been observed for some tumour types but not all (Table 3). Ultimately combination therapy is likely to be more successful. GH blockade may have application in a wide range of GH- and IGF1-responsive cancers (Kopchick et al. 2022). However, early trials should focus on a tumour type for which there is strong supporting evidence, such as breast, melanoma or HCC (Tables 1 and 2) (Perry et al. 2008, Wu et al. 2011, Sustarsic et al. 2013, Kong et al. 2016, Kaseb et al. 2022). HCC is an aggressive and difficult-to-treat cancer with limited systemic treatment options for advanced disease (Tümen et al. 2022) that might be a good candidate for clinical evaluation of GHR antagonists. Increased GH in the serum and tumours of HCC patients has been shown to correlate with poor prognosis (Tables 1 and 2), while GHR inhibition reduces HCC tumour growth in mice (Kong et al. 2016, Basu et al. 2022, Haque et al. 2022, Kaseb et al. 2022). However, confusing the issue, low GHR expression in HCC correlates with better prognosis in several recent reports (Tables 1 and 2). This highlights the need for further studies clarifying the role of GHR signalling prior to clinical applications. High serum GH in HCC may be explained by loss of negative feedback that regulates GH secretion. Kaseb et al. hypothesised that liver damage might be expected to decrease hepatic IGF1 release, which in turn leads to increased GH release from the pituitary due to the loss of negative feedback from IGF1 (Kaseb et al. 2022).

The utility of biomarkers to select patient populations for trials, to monitor toxicity and to serve as surrogate clinical efficacy endpoints will also be an important consideration. Clinical trials that use predictive biomarkers to enrich patient selection have much higher overall success rates (Wong et al. 2019). At present there is a lack of diagnostic biomarkers capable of predicting GH response in tumours and it is clear from in vitro and in vivo studies that GHR expression alone will not predict responsiveness to GHR antagonism (Divisova et al. 2006, Chiesa et al. 2011). In xenograft experiments, pegvisomant does not inhibit all tumour types (Table 3), despite expression of the GHR. Reduction in serum IGF1 measurements work very well as a surrogate biomarker for confirming inhibition of systemic GH in patients, but this does not measure tumoural GHR inhibition so other variables may need to be considered for predictive biomarkers.

Will suppression of GH signalling reduce cancer incidence in humans?

Life expectancy has increased over the last two centuries, but this has not been accompanied by a comparable increase in healthspan (the period of life free from disease) due to the increase in non-communicable diseases afflicting a growing older population (Garmany et al. 2021, Le Couteur & Barzilai 2022). Inhibition of the GH/IGF1 axis has been highlighted as a key strategy to improve human healthspan by reducing the incidence of chronic age-related diseases such as cancer and diabetes (Berryman et al. 2008, Longo et al. 2015, van der Spoel et al. 2016, Aguiar-Oliveira & Bartke 2019, Lu et al. 2019, Duran-Ortiz et al. 2021, Bartke 2022, Brown-Borg 2022). Supporting this, humans born with Laron syndrome have remarkably reduced incidence of cancer, increased insulin sensitivity and youthful cognitive and motor functions (Laron et al. 2017, Guevara-Aguirre et al. 2020, Werner et al. 2020). Animal models of Laron syndrome or GH insensitivity also have reduced cancer rates and are protected from experimentally induced tumours (Bartke et al. 2016, Duran-Ortiz et al. 2021). Mice with postnatal deletion of the Ghr gene from 6 weeks also appear to have many of the beneficial effects observed in long-lived Ghr knockout mice (Junnila et al. 2016). Beneficial effects are also seen with Ghr gene disruption in mature animals, with sexually dimorphic effects observed on lifespan and cancer (Duran-Ortiz et al. 2021). While pharmacological suppression of GH signalling has been proposed as a means of protecting against cancer, this field is still in its infancy and the potential treatment window critical for extended healthspan is unclear. Ongoing investigations in this area will be of interest.

Conclusion

There is considerable evidence that GH signalling through the GHR can promote cancer progression; however, although several GHR antagonists have been developed, there is little known about how to optimise their use in cancer patients in the clinic. Identifying biomarkers for patient selection and efficacy endpoint monitoring will be critical, with additional preclinical studies required. For preclinical models to effectively predict clinical activity, therapies will need to block both human and mouse GH signalling. Development of a surrogate rodent-specific Ghr inhibitor is therefore recommended. Ultimately, an increased interest in preclinical drug development will expand the options available for clinical and research applications and can inform large-scale clinical studies where GH and GHR expression in tumours, circulating GH, clinicopathological features and survival outcome are profiled in patients to identify optimal use of these agents.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This work did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

References

  • Abu El-Makarem MA, Kamel MF, Mohamed AA, Ali HA, Mohamed MR, Mohamed AE-DM, El-Said AM, Ameen MG, Hassnine AA & & Hassan HA 2022 Down-regulation of hepatic expression of GHR/STAT5/IGF-1 signaling pathway fosters development and aggressiveness of HCV-related hepatocellular carcinoma: crosstalk with Snail-1 and type 2 transforming growth factor-beta receptor. PLoS One 17 e0277266. (https://doi.org/10.1371/journal.pone.0277266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aguiar-Oliveira MH & & Bartke A 2019 Growth hormone deficiency: health and longevity. Endocrine Reviews 40 575601. (https://doi.org/10.1210/er.2018-00216)

  • Ariyasu D, Kubo E, Higa D, Shibata S, Takaoka Y, Sugimoto M, Imaizumi K, Hasegawa T & & Araki K 2019 Decreased activity of the Ghrhr and gh promoters causes dominantly inherited GH deficiency in humanized GH1 mouse models. Endocrinology 160 26732691. (https://doi.org/10.1210/EN.2019-00306)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arumugam A, Subramani R, Nandy SB, Terreros D, Dwivedi AK, Saltzstein E & & Lakshmanaswamy R 2019 Silencing growth hormone receptor inhibits estrogen receptor negative breast cancer through ATP-binding cassette sub-family G member 2. Experimental and Molecular Medicine 51 113. (https://doi.org/10.1038/s12276-018-0197-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A & & Kopchick JJ 2015 The forgotten lactogenic activity of growth hormone: important implications for rodent studies. Endocrinology 156 16201622. (https://doi.org/10.1210/en.2015-1097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A, List EO & & Kopchick JJ 2016 The somatotropic axis and aging: benefits of endocrine defects. Growth Hormone and IGF Research 27 4145. (https://doi.org/10.1016/j.ghir.2016.02.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A 2022 Somatotropic axis, pace of life and aging. Frontiers in Endocrinology 13 916139. (https://doi.org/10.3389/fendo.2022.916139)

  • Basu R & & Kopchick JJ 2019 The effects of growth hormone on therapy resistance in cancer. Cancer Drug Resistance 2 827846. (https://doi.org/10.20517/cdr.2019.27)

  • Basu R, Nahar K, Kulkarni P, Kerekes O, Sattler M, Hall Z, Neggers S, Holub JM & & Kopchick JJ 2021 A novel peptide antagonist of the human growth hormone receptor. Journal of Biological Chemistry 296 100588. (https://doi.org/10.1016/j.jbc.2021.100588)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Basu R, Qian Y, Mathes S, Terry J, Arnett N, Riddell T, Stevens A, Funk K, Bell S, Bokal Z, et al.2022 Growth hormone receptor antagonism downregulates ATP-binding cassette transporters contributing to improved drug efficacy against melanoma and hepatocarcinoma in vivo. Frontiers in Oncology 12 936145. (https://doi.org/10.3389/fonc.2022.936145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berryman DE, Christiansen JS, Johannsson G, Thorner MO & & Kopchick JJ 2008 Role of the GH/IGF-1 axis in lifespan and healthspan: lessons from animal models. Growth Hormone and IGF Research 18 455471. (https://doi.org/10.1016/j.ghir.2008.05.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Borcherding DC, Hugo ER, Fox SR, Jacobson EM, Hunt BG, Merino EJ & & Ben-Jonathan N 2021 Suppression of breast cancer by small molecules that block the prolactin receptor. Cancers 13. (https://doi.org/10.3390/cancers13112662)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brittain AL, Basu R, Qian Y & & Kopchick JJ 2017 Growth hormone and the epithelial-to-mesenchymal transition. Journal of Clinical Endocrinology and Metabolism 102 36623673. (https://doi.org/10.1210/jc.2017-01000)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brody R, Zupancic T, Kopchick J & & Basu R 2020 Therapeutic Pegylated Growth Hormone Antagonists, US20200390863A1.. US Patent. (available at: https://patents.google.com/patent/US11452763B2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brody R, ZupancVic T, Sandbhor U, Kopchick J & & Basu R 2021 Growth Hormone Antagonist and Anti-cancer Composition Combination Therapy, US20220040266A1. US Patent. (available at: https://patents.google.com/patent/WO2023028430A3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brooks AJ, Dai W, O’Mara ML, Abankwa D, Chhabra Y, Pelekanos RA, Gardon O, Tunny KA, Blucher KM, Morton CJ, et al.2014 Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science 344 1249783. (https://doi.org/10.1126/science.1249783)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brown-Borg HM 2022 Growth hormone, not IGF-1 is the key longevity regulator in mammals. Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 77 17191723. (https://doi.org/10.1093/GERONA/GLAC092)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulatowicz JJ & & Wood TL 2022 Activation versus inhibition of IGF1R: a dual role in breast tumorigenesis. Frontiers in Endocrinology 13 911079. (https://doi.org/10.3389/FENDO.2022.911079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cai R, Zhang X, Wang H, Cui T, Halmos G, Sha W, He J, Popovics P, Vidaurre I, Zhang C, et al.2022a Synthesis of potent antagonists of receptors for growth hormone-releasing hormone with antitumor and anti-inflammatory activity. Peptides 150 170716. (https://doi.org/10.1016/j.peptides.2021.170716)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cai D, Zhao Z, Hu J, Dai X, Zhong G, Gong J & & Qi F 2022b Identification of the tumor immune microenvironment and therapeutic biomarkers by a novel molecular subtype based on aging-related genes in hepatocellular carcinoma. Frontiers in Surgery 9 836080. (https://doi.org/10.3389/fsurg.2022.836080)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cao J & & Yee D 2021 Disrupting insulin and IGF receptor function in cancer. International Journal of Molecular Sciences 22 113. (https://doi.org/10.3390/ijms22020555)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang TC, Lin JJ, Yu SC & & Chang TJ 1990 Absence of growth-hormone receptor in hepatocellular carcinoma and cirrhotic liver. Hepatology 11 123126. (https://doi.org/10.1002/hep.1840110121)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen X, Wu D, Zheng Y, Liu X & & Wang J 2020 Preparation of a growth hormone receptor/prolactin receptor bispecific antibody antagonist which exhibited anti-cancer activity. Frontiers in Pharmacology 11 598423. (https://doi.org/10.3389/fphar.2020.598423)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng Y, Li W, Gui R, Wang C, Song J, Wang Z, Wang X, Shen Y, Wang Z & & Hao L 2021 Dual characters of GH-IGF1 signaling pathways in radiotherapy and post-radiotherapy repair of cancers. Frontiers in Cell and Developmental Biology 9 671247. (https://doi.org/10.3389/fcell.2021.671247)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chesnokova V & & Melmed S 2019 Growth hormone in the tumor microenvironment. Archives of Endocrinology and Metabolism 63 568575. (https://doi.org/10.20945/2359-3997000000186)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chhabra Y, Waters MJ & & Brooks AJ 2011 Role of the growth hormone–IGF-1 axis in cancer. Expert Review of Endocrinology and Metabolism 6 7184. (https://doi.org/10.1586/eem.10.73)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chhabra Y, Wong HY, Nikolajsen LF, Steinocher H, Papadopulos A, Tunny KA, Meunier FA, Smith AG, Kragelund BB, Brooks AJ, et al.2018 A growth hormone receptor SNP promotes lung cancer by impairment of SOCS2-mediated degradation. Oncogene 37 489501. (https://doi.org/10.1038/onc.2017.352)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chiesa J, Ferrer C, Arnould C, Vouyovitch CM, Diaz JJ, Gonzalez S, Mares P, Morel G, Wu ZS, Zhu T, et al.2011 Autocrine proliferative effects of hGH are maintained in primary cultures of human mammary carcinoma cells. Journal of Clinical Endocrinology and Metabolism 96 E1418E1426. (https://doi.org/10.1210/jc.2011-0473)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Costantini VJA, Vicentini E, Sabbatini FM, Valerio E, Lepore S, Tessari M, Sartori M, Michielin F, Melotto S, Bifone A, et al.2011 GSK1614343, a novel ghrelin receptor antagonist, produces an unexpected increase of food intake and body weight in rodents and dogs. Neuroendocrinology 94 158168. (https://doi.org/10.1159/000328968)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dabrosin N & & Dabrosin C 2020 Postmenopausal dense breasts maintain premenopausal levels of GH and insulin-like growth factor binding proteins in vivo. Journal of Clinical Endocrinology and Metabolism 105 16171628. (https://doi.org/10.1210/CLINEM/DGZ323)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dagnaes-Hansen F, Duan H, Rasmussen LM, Friend KE & & Flyvbjerg A 2004 Growth hormone receptor antagonist administration inhibits growth of human colorectal carcinoma in nude mice. Anticancer Research 24 37353742.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Divisova J, Kuiatse I, Lazard Z, Weiss H, Vreeland F, Hadsell DL, Schiff R, Osborne CK & & Lee v AV 2006 The growth hormone receptor antagonist pegvisomant blocks both mammary gland development and MCF-7 breast cancer xenograft growth. Breast Cancer Research and Treatment 98 315327. (https://doi.org/10.1007/s10549-006-9168-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duran-Ortiz S, List EO, Basu R & & Kopchick JJ 2021 Extending lifespan by modulating the growth hormone/insulin-like growth factor-1 axis: coming of age. Pituitary 24 438456. (https://doi.org/10.1007/s11102-020-01117-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Emerman JT, Leahy M, Gout PW & & Bruchovsky N 1985 Elevated growth hormone levels in sera from breast cancer patients. Hormone and Metabolic Research 17 421424. (https://doi.org/10.1055/s-2007-1013563)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Evans A, Jamieson SMF, Liu DX, Wilson WR & & Perry JK 2016 Growth hormone receptor antagonism suppresses tumour regrowth after radiotherapy in an endometrial cancer xenograft model. Cancer Letters 379 117123. (https://doi.org/10.1016/j.canlet.2016.05.031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fodor D, Pozsgai É, Schally AV, László Z, Gömöri É, Szabó É, Rumi L, Lőcsei D, Boronkai Á & & Bellyei S 2023 Expression levels of GHRH-receptor, pAkt and Hsp90 predict 10-year overall survival in patients with locally advanced rectal cancer. Biomedicines 11 719. (https://doi.org/10.3390/biomedicines11030719)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frank SJ 2020 Classical and novel GH receptor signaling pathways. Molecular and Cellular Endocrinology 518 110999. (https://doi.org/10.1016/J.MCE.2020.110999)

  • Gao CM, Ding JH, Wu JS, Cao HX, Li SP, Liu YT, Tang JH & & Tajima K 2015 Growth hormone 1 T1663A polymorphism, recreational physical activity and BMI, and breast cancer risk in Chinese women. Asian Pacific Journal of Cancer Prevention 16 54215425. (https://doi.org/10.7314/APJCP.2015.16.13.5421)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • García-Caballero T, Mertani HM, Lambert A, Gallego R, Fraga M, Pintos E, Forteza J, Chevallier M, Lobie PE, Vonderhaar BK, et al.2000 Increased expression of growth hormone and prolactin receptors in hepatocellular carcinomas. Endocrine 12 265271. (https://doi.org/10.1385/ENDO:12:3:265)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garmany A, Yamada S & & Terzic A 2021 Longevity leap: mind the healthspan gap. NPJ Regenerative Medicine 6 57. (https://doi.org/10.1038/s41536-021-00169-5)

  • Gebre-Medhin M, Kindblom LG, Wennbo H, Törnell J & & Meis-Kindblom JM 2001 Growth hormone receptor is expressed in human breast cancer. American Journal of Pathology 158 12171222. (https://doi.org/10.1016/S0002-9440(1064071-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goffin V, Bernichtein S, Carrière O, Bennett WF, Kopchick JJ & & Kelly PA 1999 The human growth hormone antagonist B2036 does not interact with the prolactin receptor. Endocrinology 140 38533856. (https://doi.org/10.1210/endo.140.8.7047)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gross JD, Zhou Y, Barak LS & & Caron MG 2023 Ghrelin receptor signaling in health and disease: a biased view. Trends in Endocrinology and Metabolism: TEM 34 106118. (https://doi.org/10.1016/J.TEM.2022.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guevara-Aguirre J, Bautista C, Torres C, Peña G, Guevara C, Palacios C, Guevara A & & Gavilanes AWD 2020 Insights from the clinical phenotype of subjects with Laron syndrome in Ecuador. Reviews in Endocrine and Metabolic Disorders 22 5970. (https://doi.org/10.1007/s11154-020-09602-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo Y, Yang J, Gao H, Tian X, Zhang X & & Kan Q 2022 Development and verification of a combined immune- and metabolism-related prognostic signature for hepatocellular carcinoma. Frontiers in Immunology 13 927635. (https://doi.org/10.3389/fimmu.2022.927635)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamann M, Grill S, Struck J, Bergmann A, Hartmann O, Pölcher M & & Kiechle M 2019 Detection of early breast cancer beyond mammographic screening: a promising biomarker panel. Biomarkers in Medicine 13 11071117. (https://doi.org/10.2217/BMM-2019-0085)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haque A, Sahu V, Lombardo JL, Xiao L, George B, Wolff RA, Morris JS, Rashid A, Kopchick JJ, Kaseb AO, et al.2022 Disruption of growth hormone receptor signaling abrogates hepatocellular carcinoma development. Journal of Hepatocellular Carcinoma 9 823837. (https://doi.org/10.2147/JHC.S368208)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harvey S, Martínez-Moreno CG, Luna M & & Arámburo C 2015 Autocrine/paracrine roles of extrapituitary growth hormone and prolactin in health and disease: an overview. General and Comparative Endocrinology 220 103111. (https://doi.org/10.1016/j.ygcen.2014.11.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hasegawa Y, Fujii K, Yamada M, Igarashi Y, Tachibana K, Tanaka T, Onigata K, Nishi Y, Kato S & & Hasegawa T 2000 Identification of novel human GH-1 gene polymorphisms that are associated with growth hormone secretion and height. Journal of Clinical Endocrinology and Metabolism 85 12901295. (https://doi.org/10.1210/jcem.85.3.6468)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haslam A, Olivier T, Powell K, Tuia J & & Prasad V 2023 Eventual success rate and predictors of success for oncology drugs tested in phase I trials. International Journal of Cancer 152 276282. (https://doi.org/10.1002/ijc.34181)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hassouna R, Labarthe A, Zizzari P, Videau C, Culler M, Epelbaum J & & Tolle V 2013 Actions of agonists and antagonists of the ghrelin/GHS-R pathway on GH secretion, appetite, and cFos activity. Frontiers in Endocrinology 4 25. (https://doi.org/10.3389/FENDO.2013.00025)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He S, Qiao J, Wang L & & Yu L 2022 A novel immune-related gene signature predicts the prognosis of hepatocellular carcinoma. Frontiers in Oncology 12 955192. (https://doi.org/10.3389/fonc.2022.955192)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jain L, Fadason T, Schierding W, Vickers MH, O’Sullivan JM & & Perry JK 2020 3D interactions with the growth hormone locus in cellular signalling and cancer-related pathways. Journal of Molecular Endocrinology 64 209222. (https://doi.org/10.1530/JME-20-0010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jen J, LaBadie RR, Liang Y, Crownover PH, Gao X & & Hey-Hadavi JH 2013 Pegvisomant bioavailability of single 30 mg/mL subcutaneous injection compared to two 15 mg/mL subcutaneous injections: a pharmacokinetic, safety and tolerability study. Growth Hormone and IGF Research 23 114119. (https://doi.org/10.1016/j.ghir.2013.04.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang J, Wan Y, Wang X, Xu J, Harris JM, Lobie PE, Zhang Y, Zinn KR, Waters MJ & & Frank SJ 2011 Inhibitory GH receptor extracellular domain monoclonal antibodies: three-dimensional epitope mapping. Endocrinology 152 47774788. (https://doi.org/10.1210/en.2011-1336)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Junnila RK, Duran-Ortiz S, Suer O, Sustarsic EG, Berryman DE, List EO & & Kopchick JJ 2016 Disruption of the GH receptor gene in adult mice increases maximal lifespan in females. Endocrinology 157 45024513. (https://doi.org/10.1210/en.2016-1649)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaseb AO, Haque A, Vishwamitra D, Hassan MM, Xiao L, George B, Sahu V, Mohamed YI, Carmagnani Pestana R, Lombardo JL, et al.2022 Blockade of growth hormone receptor signaling by using pegvisomant: a functional therapeutic strategy in hepatocellular carcinoma. Frontiers in Oncology 12 986305. (https://doi.org/10.3389/fonc.2022.986305)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaulsay KK, Mertani HC, Törnell J, Morel G, Lee KO & & Lobie PE 1999 Autocrine stimulation of human mammary carcinoma cell proliferation by human growth hormone. Experimental Cell Research 250 3550. (https://doi.org/10.1006/excr.1999.4492)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaulsay KK, Zhu T, Bennett WF, Lee KO & & Lobie PE 2001 The effects of autocrine human growth hormone (hGH) on human mammary carcinoma cell behavior are mediated via the hGH receptor. Endocrinology 142 767777. (https://doi.org/10.1210/endo.142.2.7936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kawasaki K, Rekhtman N, Quintanal-Villalonga Á & & Rudin CM 2023 Neuroendocrine neoplasms of the lung and gastrointestinal system: convergent biology and a path to better therapies. Nature Reviews. Clinical Oncology 20 1632. (https://doi.org/10.1038/s41571-022-00696-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khoury-Shakour S, Gruber SB, Lejbkowicz F, Rennert HS, Raskin L, Pinchev M & & Rennert G 2008 Recreational physical activity modifies the association between a common GH1 polymorphism and colorectal cancer risk. Cancer Epidemiology, Biomarkers and Prevention 17 33143318. (https://doi.org/10.1158/1055-9965.EPI-08-0062)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kodama T, Kodama M, Jenkins NA, Copeland NG, Chen HJ & & Wei Z 2022 Ring finger Protein 125 is an anti-proliferative tumor suppressor in hepatocellular carcinoma. Cancers 14. (https://doi.org/10.3390/CANCERS14112589)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kong X, Wu W, Yuan Y, Pandey V, Wu Z, Lu X, Zhang W, Chen Y, Wu M, Zhang M, et al.2016 Human growth hormone and human prolactin function as autocrine/paracrine promoters of progression of hepatocellular carcinoma. Oncotarget 7 2946529479. (https://doi.org/10.18632/oncotarget.8781)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kopchick JJ, Parkinson C, Stevens EC & & Trainer PJ 2002 Growth hormone receptor antagonists: discovery, development, and use in patients with acromegaly. Endocrine Reviews 23 623646. (https://doi.org/10.1210/er.2001-0022)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kopchick JJ, List EO, Kelder B, Gosney ES & & Berryman DE 2014 Evaluation of growth hormone (GH) action in mice: discovery of GH receptor antagonists and clinical indications. Molecular and Cellular Endocrinology 386 3445. (https://doi.org/10.1016/j.mce.2013.09.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kopchick JJ, Basu R, Berryman DE, Jorgensen JOL, Johannsson G & & Puri V 2022 Covert actions of growth hormone: fibrosis, cardiovascular diseases and cancer. Nature Reviews. Endocrinology 18 558573. (https://doi.org/10.1038/s41574-022-00702-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kotta AS, Kelling AS, Corleto KA, Sun Y & & Giles ED 2022 Ghrelin and cancer: examining the roles of the ghrelin axis in tumor growth and progression. Biomolecules 12. (https://doi.org/10.3390/biom12040483)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Labadzhyan A & & Melmed S 2022 Molecular targets in acromegaly. Frontiers in Endocrinology 13 1068061. (https://doi.org/10.3389/fendo.2022.1068061)

  • Lantvit DD, Unterberger CJ, Lazar M, Arneson PD, Longhurst CA, Swanson SM & & Marker PC 2021 Mammary tumors growing in the absence of growth hormone are more sensitive to doxorubicin than wild-type tumors. Endocrinology 162. (https://doi.org/10.1210/endocr/bqab013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Laron Z, Kauli R, Lapkina L & & Werner H 2017 IGF-I deficiency, longevity and cancer protection of patients with Laron syndrome. Mutation Research. Reviews in Mutation Research 772 123133. (https://doi.org/10.1016/j.mrrev.2016.08.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Le Couteur DG & & Barzilai N 2022 New horizons in life extension, healthspan extension and exceptional longevity. Age and Ageing 51. (https://doi.org/10.1093/ageing/afac156)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • le Marchand L, Donlon T, Seifried A, Kaaks R, Rinaldi S & & Wilkens LR 2002 Association of a common polymorphism in the human GH1 gene with colorectal neoplasia. Journal of the National Cancer Institute 94 454460. (https://doi.org/10.1093/jnci/94.6.454)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leporati P, Fonte R, de Martinis L, Zambelli A, Magri F, Pavesi L, Rotondi M & & Chiovato L 2015 A male patient with acromegaly and breast cancer: treating acromegaly to control tumor progression. BMC Cancer 15 397. (https://doi.org/10.1186/s12885-015-1400-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liao S, Vickers MH, Stanley JL, Baker PN & & Perry JK 2018 Human placental growth hormone variant in pathological pregnancies. Endocrinology 159 21862198. (https://doi.org/10.1210/en.2018-00037)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin CC, Liu TW, Yeh ML, Tsai YS, Tsai PC, Huang CF, Huang JF, Chuang WL, Dai CY & & Yu ML 2021 Significant down-regulation of growth hormone receptor expression revealed as a new unfavorable prognos- tic factor in hepatitis C virus-related hepatocellular carcinoma. Clinical and Molecular Hepatology 27 313328. (https://doi.org/10.3350/cmh.2020.0247)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu JP, Wang HT, Ou QJ, Lu YT, Lu HP, Gao JH, Chu ZH & & Zhao HY 2003 Expression of growth hormone receptor in hepatocellular carcinoma and its significance. Ai Zheng 22 298301.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu Y, Zhang Y, Jiang J, Lobie PE, Paulmurugan R, Langenheim JF, Chen WY, Zinn KR & & Frank SJ 2016 GHR/PRLR heteromultimer is composed of GHR homodimers and PRLR homodimers. Molecular Endocrinology 30 504517. (https://doi.org/10.1210/me.2015-1319)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lombardi S, Honeth G, Ginestier C, Shinomiya I, Marlow R, Buchupalli B, Gazinska P, Brown J, Catchpole S, Liu S, et al.2014 Growth hormone is secreted by normal breast epithelium upon progesterone stimulation and increases proliferation of stem/progenitor cells. Stem Cell Reports 2 780793. (https://doi.org/10.1016/j.stemcr.2014.05.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Longo VD, Antebi A, Bartke A, Barzilai N, Brown-Borg HM, Caruso C, Curiel TJ, de Cabo R, Franceschi C, Gems D, et al.2015 Interventions to slow aging in humans: are we ready? Aging Cell 14 497510. (https://doi.org/10.1111/acel.12338)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Love RR, Rose DR, Surawicz TS & & Newcomb PA 1991 Prolactin and growth hormone levels in premenopausal women with breast cancer and healthy women with a strong family history of breast cancer. Cancer 68 14011405. (https://doi.org/10.1002/1097-0142(19910915)68:6<1401::AID-CNCR2820680637>3.0.CO;2-K)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu M, Flanagan JU, Langley RJ, Hay MP & & Perry JK 2019 Targeting growth hormone function: strategies and therapeutic applications. Signal Transduction and Targeted Therapy 4 3. (https://doi.org/10.1038/s41392-019-0036-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu H, Zhou Q, He J, Jiang Z, Peng C, Tong R & & Shi J 2020 Recent advances in the development of protein-protein interactions modulators: mechanisms and clinical trials. Signal Transduction and Targeted Therapy 5 213. (https://doi.org/10.1038/s41392-020-00315-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mai L, Qiu Y, Lian Z, Chen C, Wang L, Yin Y, Wang S, Yang X, Li Y, Peng W, et al.2021 MustSeq, an alternative approach for multiplexible strand-specific 3’ end sequencing of mRNA transcriptome confers high efficiency and practicality. RNA Biology 18(sup1) 232243. (https://doi.org/10.1080/15476286.2021.1974208)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Manning M, Jiang Y, Wang R, Liu L, Rode S, Bonahoom M, Kim S & & Yang ZQ 2020 Pan-cancer analysis of RNA methyltransferases identifies FTSJ3 as a potential regulator of breast cancer progression. RNA Biology 17 474486. (https://doi.org/10.1080/15476286.2019.1708549)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mazzoccoli G, Giuliani A, Bianco G, De Cata A, Balzanelli M, Carella AM, La Viola M & & Tarquini R 1999 Decreased serum levels of insulin-like growth factor (IGF)-I in patients with lung cancer: temporal relationship with growth hormone (GH) levels. Anticancer Research 19 13971399.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McCutcheon IE, Flyvbjerg A, Hill H, Li J, Bennett WF, Scarlett JA & & Friend KE 2001 Antitumor activity of the growth hormone receptor antagonist pegvisomant against human meningiomas in nude mice. Journal of Neurosurgery 94 487492. (https://doi.org/10.3171/jns.2001.94.3.0487)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Melmed S, Bronstein MD, Chanson P, Klibanski A, Casanueva FF, Wass JAH, Strasburger CJ, Luger A, Clemmons DR & & Giustina A 2018 A Consensus Statement on acromegaly therapeutic outcomes. Nature Reviews. Endocrinology 14 552561. (https://doi.org/10.1038/s41574-018-0058-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Menashe I, Maeder D, Garcia-Closas M, Figueroa JD, Bhattacharjee S, Rotunno M, Kraft P, Hunter DJ, Chanock SJ, Rosenberg PS, et al.2010 Pathway analysis of breast cancer genome-wide association study highlights three pathways and one canonical signaling cascade. Cancer Research 70 44534459. (https://doi.org/10.1158/0008-5472.CAN-09-4502)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meng Y, Zhou B, Pei Z, Chen Y & & Chang D 2022 The nuclear-localized GHR is involved in the cell proliferation of gastric cancer, and pegvisomant may be an important potential drug to inhibit the proliferation of gastric cancer cells. Biochemistry and Cell Biology 100 125135. (https://doi.org/10.1139/bcb-2021-0386)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mertani HC, Garcia-Caballero T, Lambert A, Gérard F, Palayer C, Boutin JM, Vonderhaar BK, Waters MJ, Lobie PE & & Morel G 1998 Cellular expression of growth hormone and prolactin receptors in human breast disorders. International Journal of Cancer 79 202211. (https://doi.org/10.1002/(sici)1097-0215(19980417)79:2<202::aid-ijc17>3.0.co;2-b)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Millar DS, Horan M, Chuzhanova NA & & Cooper DN 2010 Characterisation of a functional intronic polymorphism in the human growth hormone (GH1) gene. Human Genomics 4 289301. (https://doi.org/10.1186/1479-7364-4-5-289)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mohamed YI, Duda DG, Awiwi MO, Lee SS, Altameemi L, Xiao L, Morris JS, Wolff RA, Elsayes KM, Hatia RI, et al.2022 Plasma growth hormone is a potential biomarker of response to atezolizumab and bevacizumab in advanced hepatocellular carcinoma patients. Oncotarget 13 13141321. (https://doi.org/10.18632/oncotarget.28322)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mol JA, Henzen-Logmans SC, Hageman P, Misdorp W, Blankenstein MA & & Rijnberk A 1995 Expression of the gene encoding growth hormone in the human mammary gland. Journal of Clinical Endocrinology and Metabolism 80 30943096. (https://doi.org/10.1210/jcem.80.10.7559904)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mordenti J, Chen SA, Moore JA, Ferraiolo BL & & Green JD 1991 Interspecies scaling of clearance and volume of distribution data for five therapeutic proteins. Pharmaceutical Research 8 13511359. (https://doi.org/10.1023/a:1015836720294)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mukhina S, Mertani HC, Guo K, Lee KO, Gluckman PD & & Lobie PE 2004 Phenotypic conversion of human mammary carcinoma cells by autocrine human growth hormone. Proceedings of the National Academy of Sciences of the United States of America 101 1516615171. (https://doi.org/10.1073/pnas.0405881101)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Neggers SJCMM, Muhammad A & & van der Lely AJ 2016 Pegvisomant treatment in acromegaly. Neuroendocrinology 103 5965. (https://doi.org/10.1159/000381644)

  • Nero TL, Morton CJ, Holien JK, Wielens J & & Parker MW 2014 Oncogenic protein interfaces: small molecules, big challenges. Nature Reviews. Cancer 14 248262. (https://doi.org/10.1038/nrc3690)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perry JK, Emerald BS, Mertani HC & & Lobie PE 2006 The oncogenic potential of growth hormone. Growth Hormone and IGF Research 16 277289. (https://doi.org/10.1016/j.ghir.2006.09.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perry JK, Mohankumar KM, Emerald BS, Mertani HC & & Lobie PE 2008 The contribution of growth hormone to mammary neoplasia. Journal of Mammary Gland Biology and Neoplasia 13 131145. (https://doi.org/10.1007/s10911-008-9070-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perry JK, Liu DX, Wu ZS, Zhu T & & Lobie PE 2013 Growth hormone and cancer: an update on progress. Current Opinion in Endocrinology, Diabetes, and Obesity 20 307313. (https://doi.org/10.1097/MED.0b013e328363183a)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perry JK, Wu ZS, Mertani HC, Zhu T & & Lobie PE 2017 Tumour-derived human growth hormone as a therapeutic target in oncology. Trends in Endocrinology and Metabolism 28 587596. (https://doi.org/10.1016/j.tem.2017.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Price ML, Ley CD & & Gorvin CM 2021 The emerging role of heterodimerisation and interacting proteins in ghrelin receptor function. Journal of Endocrinology 252 R23–R39. (https://doi.org/10.1530/JOE-21-0206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pudjihartono M, Perry JK, Print C, O’Sullivan JM & & Schierding W 2022 Interpretation of the role of germline and somatic non-coding mutations in cancer: expression and chromatin conformation informed analysis. Clinical Epigenetics 14 120. (https://doi.org/10.1186/s13148-022-01342-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qian Y, Basu R, Mathes SC, Arnett NA, Duran-Ortiz S, Funk KR, Brittain AL, Kulkarni P, Terry JC, Davis E, et al.2020 Growth hormone upregulates mediators of melanoma drug efflux and epithelial-to-mesenchymal transition in vitro and in vivo. Cancers 12 126. (https://doi.org/10.3390/CANCERS12123640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Raccurt M, Lobie PE, Moudilou E, Garcia-Caballero T, Frappart L, Morel G & & Mertani HC 2002 High stromal and epithelial human gh gene expression is associated with proliferative disorders of the mammary gland. Journal of Endocrinology 175 307318. (https://doi.org/10.1677/JOE.0.1750307)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rai U, Thrimawithana TR, Valery C & & Young SA 2015 Therapeutic uses of somatostatin and its analogues: current view and potential applications. Pharmacology and Therapeutics 152 98110. (https://doi.org/10.1016/j.pharmthera.2015.05.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Recouvreux MV, Wu JB, Gao AC, Zonis S, Chesnokova V, Bhowmick N, Chung LW & & Melmed S 2017 Androgen receptor regulation of local growth hormone in prostate cancer cells. Endocrinology 158 22552268. (https://doi.org/10.1210/en.2016-1939)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rosengren L, Parrow V, Chmielewska J, Mode A & & Fhölenhag K 2007 In vivo evaluation of a novel, orally bioavailable, small molecule growth hormone receptor antagonist. Growth Hormone and IGF Research 17 4753. (https://doi.org/10.1016/j.ghir.2006.10.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rudd MF, Webb EL, Matakidou A, Sellick GS, Williams RD, Bridle H, Eisen T, Houlston RS & GELCAPS Consortium 2006 Variants in the GH-IGF axis confer susceptibility to lung cancer. Genome Research 16 693701. (https://doi.org/10.1101/gr.5120106)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sanyal A, Lajoie BR, Jain G & & Dekker J 2012 The long-range interaction landscape of gene promoters. Nature 489 109113. (https://doi.org/10.1038/nature11279)

  • Schally AV & & Varga JL 2006 Antagonists of growth hormone-releasing hormone in oncology. Combinatorial Chemistry and High Throughput Screening 9 163170. (https://doi.org/10.2174/138620706776055449)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schally AV, Varga JL & & Engel JB 2008 Antagonists of growth-hormone-releasing hormone: an emerging new therapy for cancer. Nature Clinical Practice. Endocrinology and Metabolism 4 3343. (https://doi.org/10.1038/ncpendmet0677)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi J, Tong JH & & Cai S 2014 GH1 T1663A polymorphism and cancer risk: a meta-analysis of case-control studies. Tumour Biology 35 45294538. (https://doi.org/10.1007/s13277-013-1596-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Siebel S, Rocha S, Chaparro-Riggers J, Pascua E, Lindquist K, Dilley J, Melton Z, Camacho-Hubner C & & Lin J 2014 Humanization and characterization of a human growth hormone receptor (GHR) antagonist antibody RN172. Endocrine Reviews 35.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Soleyman-Jahi S, Sadeghi F, Pastaki Khoshbin A, Khani L, Roosta V & & Zendehdel K 2019 Attribution of ghrelin to cancer; attempts to unravel an apparent controversy. Frontiers in Oncology 9 1014. (https://doi.org/10.3389/FONC.2019.01014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Souza SC, Frick GP, Wang X, Kopchick JJ, Lobo RB & & Goodman HM 1995 A single arginine residue determines species specificity of the human growth hormone receptor. Proceedings of the National Academy of Sciences of the United States of America 92 959963. (https://doi.org/10.1073/pnas.92.4.959)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Strous GJ, Almeida ADS, Putters J, Schantl J, Sedek M, Slotman JA, Nespital T, Hassink GC & & Mol JA 2020 Growth hormone receptor regulation in cancer and chronic diseases. Frontiers in Endocrinology 11 597573. (https://doi.org/10.3389/fendo.2020.597573)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun F, Liu Y, Sun H & & Tian B 2015 Development and characterization of a novel GHR antibody antagonist, GF185. International Journal of Biological Macromolecules 79 864870. (https://doi.org/10.1016/j.ijbiomac.2015.05.039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun D, Gao W, Hu H & & Zhou S 2022 Why 90% of clinical drug development fails and how to improve it? Acta Pharmaceutica Sinica. B 12 30493062. (https://doi.org/10.1016/j.apsb.2022.02.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sustarsic EG, Junnila RK & & Kopchick JJ 2013 Human metastatic melanoma cell lines express high levels of growth hormone receptor and respond to GH treatment. Biochemical and Biophysical Research Communications 441 144150. (https://doi.org/10.1016/j.bbrc.2013.10.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Swanson SM & & Unterman TG 2002 The growth hormone-deficient Spontaneous Dwarf rat is resistant to chemically induced mammary carcinogenesis. Carcinogenesis 23 977982. (https://doi.org/10.1093/CARCIN/23.6.977)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Trainer PJ, Newell-Price JDC, Ayuk J, Aylwin SJB, Rees A, Drake W, Chanson P, Brue T, Webb SM, Fajardo C, et al.2018 A randomised, open-label, parallel group phase 2 study of antisense oligonucleotide therapy in acromegaly. European Journal of Endocrinology 179 97108. (https://doi.org/10.1530/EJE-18-0138)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Triantafillidis JK, Merikas E, Govosdis V, Konstandellou E, Cheracakis P, Barbatzas C, Tzourmakliotis D & & Peros G 2003 Increased fasting serum levels of growth hormone and gastrin in patients with gastric and large bowel cancer. Hepato-Gastroenterology 50(Supplement 2) cclviccclx.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tsai YC, Cooke NE & & Liebhaber SA 2016 Long-range looping of a locus control region drives tissue-specific chromatin packing within a multigene cluster. Nucleic Acids Research 44 46514664. (https://doi.org/10.1093/nar/gkw090)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tümen D, Heumann P, Gülow K, Demirci CN, Cosma LS, Müller M & & Kandulski A 2022 Pathogenesis and current treatment strategies of hepatocellular carcinoma. Biomedicines 10. (https://doi.org/10.3390/biomedicines10123202)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Unterberger CJ, Maklakova VI, Lazar M, Arneson PD, Mcilwain SJ, Tsourkas PK, Hu R, Kopchick JJ, Swanson SM & & Marker PC 2022 GH action in prostate cancer cells promotes proliferation limits apoptosis and regulates cancer-related gene expression. Endocrinology 163. (https://doi.org/10.1210/endocr/bqac031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van den Eijnden MJ & & Strous GJ 2007 Autocrine growth hormone: effects on growth hormone receptor trafficking and signaling. Molecular Endocrinology 21 28322846. (https://doi.org/10.1210/me.2007-0092)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van der Lely AJ & & Kopchick JJ 2006 Growth hormone receptor antagonists. Neuroendocrinology 83 264268. (https://doi.org/10.1159/000095537)

  • van der Spoel E, Jansen SW, Akintola AA, Ballieux BE, Cobbaert CM, Slagboom PE, Blauw GJ, Westendorp RGJ, Pijl H, Roelfsema F, et al.2016 Growth hormone secretion is diminished and tightly controlled in humans enriched for familial longevity. Aging Cell 15 11261131. (https://doi.org/10.1111/acel.12519)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van der Velden LM, Maas P, van Amersfoort M, Timmermans-Sprang EPM, Mensinga A, van der Vaart E, Malergue F, Viëtor H, Derksen PWB, Klumperman J, et al.2022 Small molecules to regulate the GH/IGF1 axis by inhibiting the growth hormone receptor synthesis. Frontiers in Endocrinology 13 926210. (https://doi.org/10.3389/fendo.2022.926210)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wagner K, Hemminki K, Grzybowska E, Klaes R, Burwinkel B, Bugert P, Schmutzler RK, Wappenschmidt B, Butkiewicz D, Pamula J, et al.2006 Polymorphisms in genes involved in GH1 release and their association with breast cancer risk. Carcinogenesis 27 18671875. (https://doi.org/10.1093/carcin/bgl036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wagner K, Hemminki K & & Försti A 2007 The GH1/IGF-1 axis polymorphisms and their impact on breast cancer development. Breast Cancer Research and Treatment 104 233248. (https://doi.org/10.1007/s10549-006-9411-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Y, Langley RJ, Tamshen K, Harms J, Middleditch MJ, Maynard HD, Jamieson SMF & & Perry JK 2021 Enhanced bioactivity of a human GHR antagonist generated by solid-phase site-specific pegylation. Biomacromolecules 22 299308. (https://doi.org/10.1021/acs.biomac.0c01105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Y, Langley RJ, Tamshen K, Jamieson SM, Lu M, Maynard HD & & Perry JK 2020 Long-acting human growth hormone receptor antagonists produced in E. coli and conjugated with polyethylene glycol. Bioconjugate Chemistry 31 16511660. (https://doi.org/10.1021/acs.bioconjchem.0c00208)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Waters MJ & & Brooks AJ 2015 JAK2 activation by growth hormone and other cytokines. Biochemical Journal 466 111. (https://doi.org/10.1042/BJ20141293)

  • Werner H, Sarfstein R, Nagaraj K & & Laron Z 2020 Laron syndrome research paves the way for new insights in oncological investigation. Cells 9. (https://doi.org/10.3390/cells9112446)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wilkinson IR, Pradhananga SL, Speak R, Artymiuk PJ, Sayers JR & & Ross RJ 2016 A long-acting GH receptor antagonist through fusion to GH binding protein. Scientific Reports 6 35072. (https://doi.org/10.1038/srep35072)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wolin EM 2012 The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointestinal Cancer Research: GCR 5 161168.

  • Wong CH, Siah KW & & Lo AW 2019 Estimation of clinical trial success rates and related parameters. Biostatistics 20 273286. (https://doi.org/10.1093/biostatistics/kxx069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu ZS, Yang K, Wan Y, Qian PX, Perry JK, Chiesa J, Mertani HC, Zhu T & & Lobie PE 2011 Tumor expression of human growth hormone and human prolactin predict a worse survival outcome in patients with mammary or endometrial carcinoma. Journal of Clinical Endocrinology and Metabolism 96 E1619E1629. (https://doi.org/10.1210/jc.2011-1245)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yanagi S, Sato T, Kangawa K & & Nakazato M 2018 The homeostatic force of ghrelin. Cell Metabolism 27 786804. (https://doi.org/10.1016/J.CMET.2018.02.008)

  • Yin D, Vreeland F, Schaaf LJ, Millham R, Duncan BA & & Sharma A 2007 Clinical pharmacodynamic effects of the growth hormone receptor antagonist pegvisomant: implications for cancer therapy. Clinical Cancer Research 13 10001009. (https://doi.org/10.1158/1078-0432.CCR-06-1910)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zarandi M, Cai R, Kovacs M, Popovics P, Szalontay L, Cui T, Sha W, Jaszberenyi M, Varga J, Zhang X, et al.2017 Synthesis and structure-activity studies on novel analogs of human growth hormone releasing hormone (GHRH) with enhanced inhibitory activities on tumor growth. Peptides 89 6070. (https://doi.org/10.1016/j.peptides.2017.01.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang J, Fu X, Zhang N, Wang W, Liu H, Jia Y & & Nie Y 2022 Development and validation of an eight-gene signature based predictive model to evaluate the prognosis of hepatocellular carcinoma patients: a bioinformatic study. Annals of Translational Medicine 10 524524. (https://doi.org/10.21037/atm-22-1934)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu T, Starling-Emerald B, Zhang X, Lee KO, Gluckman PD, Mertani HC & & Lobie PE 2005 Oncogenic transformation of human mammary epithelial cells by autocrine human growth hormone. Cancer Research 65 317324. (https://doi.org/10.1158/0008-5472.317.65.1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu X, Li Y, Xu G & & Fu CQ 2020 Growth hormone receptor promotes breast cancer progression via the BRAF/MEK/ERK signaling pathway. FEBS Open Bio 10 1013–1020. (https://doi.org/10.1002/2211-5463.12816)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu Y, Ren C, Yang L, Zhang Z, Gong M & & Chen K 2022 Identification and validation of the high expression of pseudogene TCAM1P in cervical cancer via integrated bioinformatics analysis. Cancer Cell International 22 17. (https://doi.org/10.1186/S12935-021-02440-7)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    Regulation of GH and therapeutic blockade. GH and IGF1 affect tumour growth through endocrine, autocrine and paracrine mechanisms. GH is secreted from the anterior pituitary with release regulated by the hypothalamic hormones, growth hormone releasing hormone (GHRH) and somatostatin (SST), and ghrelin, which is predominantly produced by the stomach. Increased circulating GH promotes release of IGF1 from the liver and other target tissues. Additional regulation is provided by a feedback loop mechanism (+ and −), with high circulating GH and IGF1 inhibiting GH secretion. GH and IGF1 can also be expressed in extra-pituitary tissues and tumours, where they have local autocrine/paracrine actions on GHR- and IGF1R-expressing tumour cells, as well as cells in the tumour microenvironment. Inhibition of GH actions can be achieved through direct targeting of the GHR and by inhibition of GHR protein production. GH actions can also be blocked indirectly by inhibitors/agonists of hormones that regulate GH release from the pituitary. *GHRH, antagonists, ghrelin antagonists, and SST analogues also directly target tumour cells expressing their respective receptors. GHBP, growth hormone-binding protein; GHR, growth hormone receptor; GHRH, growth hormone releasing hormone; IGF1, insulin-like growth factor 1; SST, somatostatin. Created with BioRender.com.

  • Abu El-Makarem MA, Kamel MF, Mohamed AA, Ali HA, Mohamed MR, Mohamed AE-DM, El-Said AM, Ameen MG, Hassnine AA & & Hassan HA 2022 Down-regulation of hepatic expression of GHR/STAT5/IGF-1 signaling pathway fosters development and aggressiveness of HCV-related hepatocellular carcinoma: crosstalk with Snail-1 and type 2 transforming growth factor-beta receptor. PLoS One 17 e0277266. (https://doi.org/10.1371/journal.pone.0277266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aguiar-Oliveira MH & & Bartke A 2019 Growth hormone deficiency: health and longevity. Endocrine Reviews 40 575601. (https://doi.org/10.1210/er.2018-00216)

  • Ariyasu D, Kubo E, Higa D, Shibata S, Takaoka Y, Sugimoto M, Imaizumi K, Hasegawa T & & Araki K 2019 Decreased activity of the Ghrhr and gh promoters causes dominantly inherited GH deficiency in humanized GH1 mouse models. Endocrinology 160 26732691. (https://doi.org/10.1210/EN.2019-00306)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arumugam A, Subramani R, Nandy SB, Terreros D, Dwivedi AK, Saltzstein E & & Lakshmanaswamy R 2019 Silencing growth hormone receptor inhibits estrogen receptor negative breast cancer through ATP-binding cassette sub-family G member 2. Experimental and Molecular Medicine 51 113. (https://doi.org/10.1038/s12276-018-0197-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A & & Kopchick JJ 2015 The forgotten lactogenic activity of growth hormone: important implications for rodent studies. Endocrinology 156 16201622. (https://doi.org/10.1210/en.2015-1097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A, List EO & & Kopchick JJ 2016 The somatotropic axis and aging: benefits of endocrine defects. Growth Hormone and IGF Research 27 4145. (https://doi.org/10.1016/j.ghir.2016.02.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartke A 2022 Somatotropic axis, pace of life and aging. Frontiers in Endocrinology 13 916139. (https://doi.org/10.3389/fendo.2022.916139)

  • Basu R & & Kopchick JJ 2019 The effects of growth hormone on therapy resistance in cancer. Cancer Drug Resistance 2 827846. (https://doi.org/10.20517/cdr.2019.27)