tropomyosin receptor kinase: a novel target in screened … · v heinzle et al tropomyosin receptor...

14
https://doi.org/10.1530/ERC-17-0201 http://erc.endocrinology-journals.org © 2018 Society for Endocrinology Printed in Great Britain Published by Bioscientifica Ltd. 25:5 Endocrine-Related Cancer E T Aristizabal Prada, V Heinzle et al. Tropomyosin receptor kinase (Trk) in NETs 547–560 RESEARCH Tropomyosin receptor kinase: a novel target in screened neuroendocrine tumors Elke Tatjana Aristizabal Prada 1,2,3, *, Vera Heinzle 1,2, *, Thomas Knösel 1,4 , Svenja Nölting 1,2,3 , Gerald Spöttl 1,2,3 , Julian Maurer 1,2,3 , Christine Spitzweg 1,2,3 , Martin Angele 1,5 , Nina Schmidt 1,2,3 , Felix Beuschlein 3,6 , Günter K Stalla 7 , Rainer Blaser 8 , Klaus A Kuhn 8 and Christoph J Auernhammer 1,2,3 1 Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany 2 Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany 3 Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany 4 Institute of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany 5 Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University-Hospital, Klinikum der Universität München, Ludwig- Maximilians-University of Munich, Munich, Germany 6 Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland 7 Clinical Neuroendocrinology, Max Planck Institute of Psychiatry, Munich, Germany 8 Institute of Medical Statistics and Epidemiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany Correspondence should be addressed to C J Auernhammer: [email protected] *(E T Aristizabal Prada and V Heinzle contributed equally to this work) Abstract Tropomyosin receptor kinase (Trk) inhibitors are investigated as a novel targeted therapy in various cancers. We investigated the in vitro effects of the pan-Trk inhibitor GNF-5837 in human neuroendocrine tumor (NET) cells. The human neuroendocrine pancreatic BON1, bronchopulmonary NCI-H727 and ileal GOT1 cell lines were treated with GNF-5837 alone and in combination with everolimus. Cell viability decreased in a time- and dose- dependent manner in GOT1 cells in response to GNF-5837 treatment, while treatment in BON1 and NCI-H727 cells showed no effect on cellular viability. Trk receptor expression determined GNF-5837 sensitivity. GNF-5837 caused downregulation of PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling, the cell cycle and increased apoptotic cell death. The combinational treatment of GNF-5837 with everolimus showed a significant enhancement in inhibition of cell viability vs single substance treatments, due to a cooperative PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation, as well as an enhanced cell cycle component downregulation. Immunohistochemical staining for Trk receptors were performed using a tissue microarray containing 107 tumor samples of gastroenteropancreatic NETs. Immunohistochemical staining with TrkA receptor and pan-Trk receptor antibodies revealed a positive staining in pancreatic NETs in 24.2% (8/33) and 33.3% (11/33), respectively. We demonstrated that the pan-Trk inhibitor GNF-5837 has promising anti-tumoral properties in human NET cell lines expressing the TrkA receptor. Immunohistochemical or molecular screening for Trk expression particularly in pancreatic NETs might serve as predictive marker for molecular targeted therapy with Trk inhibitors. Key Words f tropomyosin kinase receptor (Trk) f Trk inhibitor f neuroendocrine tumor f everolimus Endocrine-Related Cancer (2018) 25, 547–560 Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AM via free access

Upload: others

Post on 06-Mar-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

https://doi.org/10.1530/ERC-17-0201http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

25:5Endocrine-Related Cancer

E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

547–560

10.1530/ERC-17-0201

RESEARCH

Tropomyosin receptor kinase: a novel target in screened neuroendocrine tumors

Elke Tatjana Aristizabal Prada1,2,3,*, Vera Heinzle1,2,*, Thomas Knösel1,4, Svenja Nölting1,2,3, Gerald Spöttl1,2,3, Julian Maurer1,2,3, Christine Spitzweg1,2,3, Martin Angele1,5, Nina Schmidt1,2,3, Felix Beuschlein3,6, Günter K Stalla7, Rainer Blaser8, Klaus A Kuhn8 and Christoph J Auernhammer1,2,3

1Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany2Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany3Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany4Institute of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany5Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany6Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland7Clinical Neuroendocrinology, Max Planck Institute of Psychiatry, Munich, Germany8Institute of Medical Statistics and Epidemiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany

Correspondence should be addressed to C J Auernhammer: [email protected]

*(E T Aristizabal Prada and V Heinzle contributed equally to this work)

Abstract

Tropomyosin receptor kinase (Trk) inhibitors are investigated as a novel targeted therapy

in various cancers. We investigated the in vitro effects of the pan-Trk inhibitor GNF-5837

in human neuroendocrine tumor (NET) cells. The human neuroendocrine pancreatic

BON1, bronchopulmonary NCI-H727 and ileal GOT1 cell lines were treated with GNF-5837

alone and in combination with everolimus. Cell viability decreased in a time- and dose-

dependent manner in GOT1 cells in response to GNF-5837 treatment, while treatment in

BON1 and NCI-H727 cells showed no effect on cellular viability. Trk receptor expression

determined GNF-5837 sensitivity. GNF-5837 caused downregulation of PI3K-Akt-mTOR

signaling, Ras-Raf-MEK-ERK signaling, the cell cycle and increased apoptotic cell death.

The combinational treatment of GNF-5837 with everolimus showed a significant

enhancement in inhibition of cell viability vs single substance treatments, due to a

cooperative PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation, as well as

an enhanced cell cycle component downregulation. Immunohistochemical staining for

Trk receptors were performed using a tissue microarray containing 107 tumor samples of

gastroenteropancreatic NETs. Immunohistochemical staining with TrkA receptor and

pan-Trk receptor antibodies revealed a positive staining in pancreatic NETs in 24.2%

(8/33) and 33.3% (11/33), respectively. We demonstrated that the pan-Trk inhibitor

GNF-5837 has promising anti-tumoral properties in human NET cell lines expressing

the TrkA receptor. Immunohistochemical or molecular screening for Trk expression

particularly in pancreatic NETs might serve as predictive marker for molecular targeted

therapy with Trk inhibitors.

5

Key Words

f tropomyosin kinase receptor (Trk)

f Trk inhibitor

f neuroendocrine tumor

f everolimus

25

Endocrine-Related Cancer (2018) 25, 547–560

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 2: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

548E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

Introduction

Neuroendocrine tumors (NETs) are highly heterogeneous tumors originating from distinct cell precursors (Schimmack et  al. 2011). Current data show an increase in incidence of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) lately (Modlin et  al. 2008a, Yao et  al. 2008). Despite increasing knowledge on the tumor biology, genetics and epigenetics of GEP-NETs, the overall outcome and survival have gained very little over the last decades (Modlin et  al. 2008b, Kidd et  al. 2016, Karpathakis et  al. 2017). Peptide receptor radionuclide therapy has shown promising results regarding progression-free and overall survival (van Vliet et al. 2013, Bodei et al. 2014), but other systemic therapeutic approaches for GEP-NETs, such as biotherapy, chemotherapy and molecular targeted therapy are limited in their efficiency (Auernhammer & Goke 2011, Pavel & Sers 2016, Cives & Strosberg 2017). Currently the only approved molecular targeting strategies for NETs are the mTORC1 inhibitor everolimus (Pavel et al. 2011, Yao et al. 2011) and the multi-tyrosin kinase inhibitor sunitinib (Raymond et al. 2011) demonstrating the urgent need for innovative molecular therapeutic targets and strategies.

As recently reviewed by Amatu et al. (2016), the Trk family TrkA, TrkB and TrkC is encoded by the NTRK genes NRTK1, NTRK2 and NTRK3, respectively. Neurotrophins (NTs) are specific ligands to the Trk receptors, especially nerve growth factor (NGF) for TrkA, brain-derived growth factor (BDGF) and NT-4/-5 for TrkB and NT-3 for TrkC (Kaplan  et  al. 1991, Klein  et  al. 1991, Barbacid 1994, Bibel & Barde 2000, Huang & Reichardt 2003, Amatu et al. 2016). All Trks are transmembrane receptors and ligand binding to the extracellular domain causes receptor oligomerization and autophosphorylation of the intracellular domain containing the kinase region. Subsequently, activated downstream effectors of the Trk signaling involve the Ras-Raf-MEK-ERK, the PI3K-Akt-mTOR, the PLCγ-1 and the PKC pathway (Huang & Reichardt 2003, Amatu et al. 2016).

The Trk receptor family has been demonstrated to be implicated in the growth of many different types of cancer, for example, oral squamous cell carcinoma (McGregor et al. 1999), pancreas (McGregor et al. 1999), thyroid (McGregor et  al. 1999), lung (Oelmann  et  al. 1995), esophagus (Zhu et al. 2000), prostate (Sortino et al. 2000), breast (Tagliabue  et  al. 2000) and ovarian cancer (Davidson et al. 2001) and glioblastoma (Oelmann et al. 1995). Chromosomal rearrangements with oncogenic gene fusions joining the 3′ region of the NTRK1, NTRK2 or NTRK3 gene to another partner gene resulting in a

constitutively activated Trk family kinase have been reported in various cancer types (Amatu et al. 2016).

Recently, Trk inhibitors have emerged as promising novel targets in molecular targeted cancer therapy (Amatu et al. 2016, Demir et al. 2016, Khotskaya et al. 2017). In a large-cell neuroendocrine carcinoma (LCNEC) cell line the pan-Trk inhibitor AZD7451 showed promising pharmacologic potential, particularly in cells with a NTRK gene fusion with constitutive Trk expression (Tatematsu et al. 2014). Furthermore, BDNF/TrkB expression was significantly upregulated in clinical LCNEC specimens and has shown to be implicated in tumorigenicity and invasiveness in vitro and in vivo (Odate et al. 2013). TrkB signaling has been associated with cellular survival and migration in neuroendocrine lung carcinomas (Osborne et al. 2013).

Several Trk inhibitors, such as LOXO-101 and entrectinib (RXDX-101), are currently investigated in clinical phase 1/2 trials and have shown encouraging activity including patients with molecularly defined cancers with rearrangements of Trk family kinases (Ardini et al. 2016, Passiglia et al. 2016, Drilon et al. 2017).

So far, no data on the putative role of Trk inhibitors in the treatment of GEP-NETs have been available. Here, we demonstrate anti-proliferative effects of the highly specific pan-Trk inhibitor GNF-5837 (Albaugh et al. 2012) in the human neuroendocrine tumor cell line GOT1 in vitro and examine TrkA downstream signal transduction. In addition, we investigated the expression profile of Trk in GEP-NET tumor samples from patients.

Materials and methods

Materials

GNF-5837 was purchased from R&D Systems and recombinant human NGF was purchased from Life Technologies. Human BDNF was obtained from Sigma-Aldrich and everolimus (07741FLUKA) was purchased from Sigma. These substances were diluted in dimethyl-sulfoxide (DMSO, 10 mM stock solution; Sigma, D8418). Dulbecco’s Modified Eagle medium–Nutrient Mixture F-12, 1:1 (DMEM/F12) as well as penicillin/streptomycin were obtained from Gibco/Invitrogen. RPMI medium (with l-Glutamine, NaCO3) and phosphate-buffered saline (PBS) were purchased from Sigma, whereas Trypsin-EDTA (10×) was acquired from PAA Laboratories (Cölbe, Deutschland). Fetal bovine serum (FBS) and amphotericin B were acquired from Biochrom (Berlin, Germany). Human adult normal brain tissue Lysate (ab29466) was obtained from Abcam and stored at −80°C.

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 3: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

549E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

Cell culture

The human pancreatic neuroendocrine BON1 tumor cell line (Evers  et al. 1991) (kindly provided by Prof. R Göke, Marburg, Germany) was grown in DMEM/F12 (1:1) supplemented with 10% FBS, 1% penicillin/streptomycin and 0.4% amphotericin B and passages (p) from p7 up to p20 were used for the main experiments. Human bronchopulmonary neuroendocrine NCI-H727 tumor cells (p11 – p23 for main experiments) (Cakir & Grossman 2011) (acquired from ATCC) and human midgut carcinoid GOT1 cells (Kolby et al. 2001) (kindly provided by Prof. O Nilsson, Sahlgrenska University Hospital Göteborg, Sweden) were grown in RPMI medium supplemented with 10% FBS, 1% penicillin/ streptomycin and 0.4% amphotericin B. The exact passage status of GOT1 cells is not known, as there is no adequate information from the original institute available. GOT1 culture medium was additionally supplemented with 5 μg/mL apo-transferrin and 0.135 IU/mL insulin. The doubling time of NET cell lines used in this study are: ≥18 day in GOT1, 42 h in NCI-H727 and 20 h in BON1 cells (Grozinsky-Glasberg et al. 2012).

All human neuroendocrine cell lines were received and cultured, as recently described (Zitzmann et al. 2007, Spampatti et al. 2014). The cells were tested mycoplasma free and incubated at 37°C and 5% CO2. All cell lines were authenticated by the German Biological Resource Centre DSMZ (DSMZ, Braunschweig, Germany) using short tandem repeats (STRs) analysis.

Cell viability assessment

Cells were counted (Cell Counter Countess, Invitrogen) seeded and grown for 24 h in 96-well plates at densities of 1500 (BON1), 2000 (NCI-H727) and 30,000 (GOT1) cells per well. The following day the cells were treated in 10% FBS medium with different concentrations of GNF-5837 alone and in combination with 10 nM everolimus, as well as with NGF. Metabolic activity was measured by ‘Cell Titer96Aqueous OneSolution’ cell viability assay (Promega) after 72-, 96- and 144-h of incubation. After 4 h of incubation with CellTiter 96solution, the absorbance at 490 nm was stated using the ELISA plate reader (Orion II, Berthold Detection Systems, Pforzheim, Germany). The highest DMSO concentration used in our experimental set up was aprox. 700 µM (0.005%).

Protein extraction and Western blotting

For Western blot experiments, 1 × 106 (GOT1) were seeded in 6-well plates and grown for 24 h in complete

medium. Then, the medium was replaced by fresh 10% FBS medium and cells were incubated with different concentrations of GNF-5837 (GNF), NGF and everolimus (RAD001). The incubation times were from 10 min up to 72 h. Short times of incubation (10 min) were needed to see the effects on Trk receptor stimulation (NGF), whereas longer times of incubation (>2 h) were needed to see the effects on Trk receptor inhibition (GNF-5837). After incubation, the cells were washed twice in cold PBS on ice and lysed in 200 μL Lysis buffer (M-PER 1 Mammalian Protein Extraction Reagent containing HALT protease & phosphatase inhibitor cocktail, Thermo Scientific). Lysates were centrifuged at 16,060 g for 10 min. Supernatants were adjusted to the same protein concentration (30–50 μg/50 μL) (Rotiquant Universal, CarlRoth, Karlsruhe, Germany) and denatured in sodium dodecyl sulfate (SDS) sample buffer (0.25% Tris–HCL, 40% glycerol, 2% SDS, 1% dithiothreitol, bromophenol blue, pH 8.8). Equal amounts of protein were separated on a SDS polyacrylamide gel and electrotransferred for 60 min onto PVDF membranes (Immobilon; Millipore) using a semi-dry Western blot technique. After blocking in 2% skimmed milk powder, the membranes were incubated overnight at 4°C in appropriate dilutions of primary antibodies against pTrkA (Tyr 674/5) 1:1000, pTrkB (Tyr 706/7) (#4621) 1:1000, TrkA (#2508) 1:1000, Trkpan (#4609) 1:1000, pAKT (Ser473) (#4060) 1:20,000, AKT(#2920) 1:10,000, pERK (Thr202/Tyr204)1/2 (#4370) 1:10,000, p4EBP1 (Ser65) (#9451) 1:2000, 4EBP1 (#9644) 1:5000; pGSK3 (Ser21/9) (#9331) 1:1000, GSK3 (#5676) 1:5000, pp70S6K (Thr389) (#9234) 1:1000, p70S6K (#9202) 1:1000, pS6 (Ser240/4) (#5364) 1:25,000, S6 (#2317) 1:2000, pEGFR (Tyr1068) (#3777) 1:1000, EGFR (#4267) 1:5000, Cyclin D3 (#2936) 1:2000 (all from Cell Signaling), Actin (A5441) 1:20,000 (Sigma), ERK1/2 (06–182) (Merck-Millipore) 1:20,000, Bcl2 (ab694) 1:1000, TrkB (ab134155) 1:1000, TrkC (ab117966) 1:500 (all from Abcam), p21 Waf1/Cip1 (BD610233) 1:2000 (BD Biosciences, Heidelberg, Germany), p53 (sc-126) 1:20000 (Santa Cruz) Anti-NGF (EP1320Y) (ab52918) 1:2000 (Abcam). After washing in TBS, the membranes were incubated with a peroxidase conjugated secondary antibody (Anti-Rabbit-IgG, HRP-linked Antibody, Cell Signaling, #7074 or Anti-Mouse-IgG, HRP-linked Antibody, Cell Signaling, #7076) 1:25,000 for 2 h. The blots were washed and immersed in the chemiluminescent substrate Super Signal West Dura (Thermo Scientific), and images were taken with an ECL ChemoCam Imager (INTAS, Göttingen, Germany).

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 4: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

550E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

Cell cycle analysis by flow cytometric analysis (FACS)

Cell cycle phase distribution was analyzed using propidium iodide staining and flow cytometry (BD Accuri C6 Analysis, BD Biosciences). Cells were cultured in 6-well plates for 24 h. Subsequently, medium was replaced by fresh medium and cells were incubated with different concentrations of GNF-5837. After 72 h, cells were washed with PBS and treated with 300 μL trypsin for 5 min at 37°C. Cells were collected, washed and resuspended in 300 μL propidium iodide solution (Sigma-Aldrich).

Patient cohort and generation of a tissue microarray (TMA)

Patients with NET of the gastroenteropancreatic system treated at the Klinikum der Universität München were enrolled in the NeoExNET (Network of Excellence for Neuroendocrine Tumors in Munich) program. The NeoExNET program was approved by the Local Ethics Committee of the University of Munich and by the data safety manager (Datenschutzbeauftragter) of the University of Munich, and all patients had given written informed consent. Paraffin-embedded tissue samples from pathology specimens of patients enrolled in the NeoExNET program were used to generate a tissue microarray (TMA) of GEP-NETs. The TMA encompassed tumor samples from 107 patients. Histology was reviewed in all tumors to confirm the diagnosis. The specimens were classified to the new WHO 2010 classification scheme and tumor grading (G) was assessed (‘G1’ = 48 patients, ‘G2’ = 51 patients, ‘G3’ = 3 patients and ‘no information’ = 5 patients). A tissue microarray was assembled using 0.6 mm punch biopsies from all samples according to standard procedures (Knosel et al. 2005, 2006).

Immunohistochemical staining of the TMA

Immunohistochemical staining was performed using TrkA (#2508) and Trkpan (#4609) 1:100 from Cell Signaling. Slides were pre-treated by cooking, and then incubated with the antibodies, followed by antibody detection using a biotinylated anti-mouse secondary antibody and a multilink biotin-streptavidin-amplified detection system (Biogenex, San Ramon, CA, USA). A Fastred chromogen system (DAKO) was used to visualize staining. A 4-tier scale was used for a semi-quantitative score (0 = negative, 1 = weak expression, 2 = moderate expression, 3 = strong expression).

Statistical analysis

The results are displayed as mean ± standard deviation of the mean (s.d.) of at least three independently performed experiments. Each cell viability experiment consisted of at least 6 samples per substance concentration and incubation period. Some a priori tests considering the normal distribution and homogeneity of variances were performed applying the Kolmogorov–Smirnov test and the Levene’s test of the SPSS statistical package SPSS (version 13.0 for Windows, SPSS (2005)). When parametric criteria were met an ANOVA comparison of means with a post hoc Tukey test or a two-tailed t-test was performed; when non-parametric criteria were met the Kruskal–Wallis followed by the Mann–Whitney test was performed. Statistical significance was assessed at P < 0.05. For the immunohistochemistry a Pearson chi-square test was performed.

Results

GNF-5837 effectively inhibits GOT1 cell growth

Cell viability assay determined a clear decrease of GOT1 cell viability in vitro in response to the pan-TRK inhibitor GNF-5837 in a time- and dose-dependent manner (Fig. 1 and Table  1). At all three tested incubation periods (48, 96 and 144 h), significant differences when compared to untreated control cells (control DMSO) were detected (P < 0.05) (Fig. 1). At a GNF-5837 concentration of 50 nM, a plateau of dose efficacy was reached, where higher doses did not yield a better effect in cell survival decrease (Fig.  1). IC20 and IC50 inhibitory concentrations of GNF-5837 in GOT1 cells were calculated (Table 1).

Anti-proliferative effect of the Trk inhibitor GNF-5837 is associated with functional Trk expression in human neuroendocrine tumor cells

GNF-5837 was also tested on the pulmonary carcinoid cell line NCI-H727 and the pancreatic neuroendocrine tumor cell line BON1, however, without any effects on cellular growth inhibition (Supplementary Fig. 1, see section on supplementary data given at the end of this article). In relation with this observed primary GNF-5837 resistance, Western blot analysis failed to detect any expression of TrkA, TrkB or TrkC receptor in BON1 and NCI-H727 cell protein extracts (Fig.  2A). In contrast, protein extracts from human midgut carcinoid GOT1 cells showed significant levels of TrkA expression (145 kDa), faint TrkC

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 5: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

551E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

expression (145 kDa) and no TrkB expression (Fig. 2A). A strong signal was also found in protein extracts derived from GOT1 cells, using a pan-Trk antibody detecting TrkA, TrkB and TrkC (Fig. 2A).

A human adult normal brain tissue lysate (ab29466 from Abcam) served as positive control for all three specific Trk receptor antibodies (Numakawa  et  al. 2010) (Fig. 2A). The different weights of Trk in the human brain tissue (90 kDa) is explained by either alternative splicing forms of Trk (Clary & Reichardt 1994) or an immature form of the receptor (Martin-Zanca et al. 1989) (Fig. 2A).

Next, we investigated the expression levels of the TrkA ligand NGF in all three cell lines. While NGF expression was detectable in human brain tissue (positive control), BON1 and NCI-H727 cells, in GOT1 cells only a low intensity band of NGF could be detected by Western blot analysis (Fig. 2B).

Demonstration of a functional TrkA receptor in GOT1 cells by NGF-mediated TrkA activation and GNF-5837-mediated TrkA inhibition

Increasing concentrations of NGF caused a dose-dependent phosphorylation of TrkA in GOT1 cells (Fig. 3A), whereas different concentrations of BDNF failed to stimulate TrkA phosphorylation (Fig. 3A), demonstrating the well-known

specificity of the ligand NGF to its receptor TrkA (Amatu et al. 2016). Incubation of GOT1 cells with NGF caused a rapid phosphorylation of TrkA receptor after 10 min, which was partially inhibited by co-incubation with the Trk inhibitor GNF-5837 (Fig.  3B). The phosphorylation status of TrkA decreased continuously with increasing concentration of the pan-TRK inhibitor GNF-5837 (5, 50 and 500 nM) (Fig. 3C). Most prominent dephosphorylation effects were reached at the highest dose of the inhibitor (Fig.  3C). All time periods of incubation showed the same dose-dependent dephosphorylation pattern of TrkA (Fig.  3C). Interestingly, after long periods of incubation (24, 48 and 72 h) with the inhibitor GNF-5837 the unphosphorylated TrkA receptor shows a compensatory upregulation (Fig. 3C).

Opposite effects of TrkA inhibitor GNF-5837 and TrkA ligand NGF on PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling, cell cycle components and anti-apoptotic Bcl2 in GOT1 cells

Recently, GNF-5837 was reported to effectively infer in Trk-driven cancer-specific cellular pathway signaling (Albaugh et al. 2012, Shargh et al. 2016). Here, we aimed to examine which downstream signaling pathways are being affected by the Trk inhibitory function of

Figure 1Cell survival of GOT1 cells with different concentrations of the pan-TRK inhibitor GNF-5837 (from 0.1 nM to 500 nM) and different times of incubation (48, 96 and 144 h). The arithmetic means and s.d. of at least three independent experiments are shown. Statistically significant different results in comparison to Control DMSO considering ***P < 0.001.

Table 1 IC20 and IC50 concentration values (in nM) of GNF-5837-mediated inhibition of GOT1 cell proliferation after 48, 96 and

144 h of incubation.

20% inhibitory concentration (IC20) of GNF-5837 in GOT1 cells after different times of incubation 48 h IC20 = 4.30 nM 96 h IC20 = 4.23 nM 144 h IC20 = 2.45 nM50% inhibitory concentration (IC50) of GNF-5837 in GOT1 cells after 144 h of incubation 144 h IC50 ~ 660 nM

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 6: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

552E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

GNF-5837 (5, 50 and 500 nM) after 24 h of incubation (Fig.  4A and Supplementary Fig.  2). One of the most important cellular signaling pathways, which is often activated during carcinogenesis, tumor proliferation and cellular survival in NETs is the PI3K-Akt-mTOR pathway (Cantley 2002, Briest & Grabowski 2014). Here, we show that key components of that pathway, such as pAkt, pp70S6k, pS6 and p4EBP1 were also clearly downregulated after treatment with the pan-Trk inhibitor GNF-5837 (Fig.  4A and Supplementary Fig.  2). Also another main pathway involved in NET tumorigenesis, the Ras-Raf-MEK-ERK pathway, was downregulated by treatment with the pan-TRK inhibitor GNF-5837, as the expression level of pErk decreased (Fig.  4A and Supplementary Fig.  2). Also the cell cycle component

Cyclin D3 and cell cycle regulator p21 were downregulated (Fig.  4A and Supplementary Fig.  2). Interestingly, the expression level of the anti-apoptotic Bcl2 also decreased, suggesting an augmentation in apoptotic cell death mechanisms (Fig. 4A and Supplementary Fig. 2).

In contrast to the effects observed during incubation with the pan-Trk inhibitor GNF-5837 (Fig.  4A and Supplementary Fig.  2), TrkA receptor stimulation with NGF lead to opposite effects regarding PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling and cell cycle regulation (Fig.  4B and Supplementary Fig.  3). Only anti-apoptotic Bcl2 expression showed no major change in expression level upon NGF stimulation (Fig.  4B and Supplementary Fig. 3).

GNF-5837 induces apoptosis and G1 cell cycle arrest

Cell cycle analysis confirmed the suggested downregulation of the cell cycle and induction of apoptosis in response to GNF-5837 treatment, by significantly increasing the number of cells in G1 and sub-G1 phases, respectively (Fig. 7).

Combined treatment with GNF-5837 and everolimus causes additive inhibitory effects on GOT1 cell growth mediated by PI3K-Akt-mTOR pathway and Cyclin D3 downregulation

GOT1 cell viability was also analyzed combining the pan-Trk inhibitor GNF-5837 with the mTOR inhibitor everolimus (Fig.  5A). The combined treatment of GNF-5837 (500 nM) with everolimus (10 nM) showed a modest but significant enhancement over single substance

Figure 2(A) Base expression level of the Trk receptors (TrkA, TrkB, TrkC and Trkpan) in neuroendocrine tumor cells BON1, NCI-H727 and GOT1 and the positive control of human adult normal brain cells. A representative blot out of at least 3 independently conducted Western blots is shown. (Only the positive control of brain tissue was analyzed less than 3 times). (B) Base expression level of the nerve growth factor (NGF) in neuroendocrine tumor cells BON1, NCI-H727 and GOT1 was evaluated by Western blot analysis. A representative blot out of three independently performed experiments is shown. (Only the positive control of brain tissue was analyzed less than 3 times).

Figure 3TrkA activation upon specific NGF stimulation and TrkA blocking upon GNF-5837 incubation in GOT1 cells. (A) A representative blot out of three of the expression lack of pTrkA after BDNF stimulation and induction of pTrkA expression by NGF stimulation in GOT1 cells is shown. GOT1 cells were treated with increasing concentrations of BDNF (0.37 nM and 3.7 nM) and NGF (0.76 nM and 7.6 nM) for 10 min. (B) Expression of pTrkA after 2 h of incubation with GNF-5837, after 10 min of incubation with NGF 7.6 nM and after combinational treatment of GNF-5837 (10 nM) and NGF (7.6 nM) in GOT1 cells. (C) Expression level of the Trkpan receptor, the TrkA receptor and its phosphorylation level after different time periods of incubation (4, 24, 48 and 72 h) with GNF-5837 (5, 50 and 500 nM). A representative blot out of three independently conducted experiments is shown.

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 7: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

553E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

treatment after 144 h of incubation (Fig. 5A). Other times of incubation (72 h or 144 h) with different concentrations of GNF-5837 (5 nM or 500 nM) could not yield additive effects with everolimus (data not shown). The additive effect in GOT1 cell eradication was attributed to an enhanced PI3K-Akt-mTOR and Cyclin D3 downregulation (Fig.  5B and Supplementary Fig.  4). Key components of the PI3K-Akt-mTOR pathway, such as pAkt, pp70S6k, pS6 and p4EBP1, as well as the cell cycle component Cyclin D3 showed an enhanced downregulation when compared to single substance treatment (Fig.  5B and Supplementary Fig. 4). The protein expression level of all other considered parameters, such as pTrkA, TrkA, pERK, p21 and Bcl2 displayed a complementary mode of action regarding anti-proliferative signaling, when compared to the respective single substance treatments (Fig.  5B and Supplementary Fig. 4).

Immunohistochemistry showed TrkA-positive staining only in pancreatic neuroendocrine tumor specimens

In order to assess the prevalence of Trk receptor expression in actual GEP-NET tumor samples, we immunohistochemically stained a tissue microarray (TMA)

with TrkA and pan-Trk antibodies (Fig. 6). We scaled the TMA probes for a semi-quantitative score (0 = negative, 1 = weak expression, 2 = moderate expression, 3 = strong expression).

In a total of 107 (100%) GEP-NET tumor specimens on the TMA, the primary tumor localization was small intestinal (SI)-NET in 65 cases (60.7%), pancreas (p)NET in 33 (30.8%), gastric NET in 1 (0.9%), colon NET in 3 (2.8%), appendix NET in 3 (2.8%) and cancer of unknown primary (CUP) in 2 (1.9%) cases (Table 2).

The antibody against TrkA stained positive (weak and moderate) exclusively in tumor samples of pancreatic primary localization as 24% (8/33) of all pNET samples stained positive for TrkA (Table 2). A statistically significant relation between primary tumor localization and TrkA/Trkpan expression level was proved.

Using a pan-Trk antibody (detecting endogenous levels of total Trk protein encompassing TrkA, TrkB and TrkC), positive IHC expression was again found in all 8 previously TrkA-positive pNET samples (Table  2), as expected. In addition, the pan-Trk antibody caused positive staining in another 3 pNET samples which had shown negative IHC expression in the previous staining with the TrkA antibody. In total, using the pan-Trk antibody 33.3% (11/33) of all pNETs stained positive for

Figure 4(A) Effects of different concentrations of GNF-5837 (5, 50 and 500) on the expression level of different components from the cell cycle, the apoptotic response and the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway after 24 h of incubation in GOT1 cells. Basal protein expression level of proteins was evaluated by Western blot analysis. A representative blot out of three independently performed experiments is shown. (B) Effects of different concentrations of NGF (0.76 nM and 7.6 nM) on the expression level of different components from the cell cycle, the apoptotic response and the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway after 24 h of incubation in GOT1 cells. Basal protein expression level of proteins was evaluated by Western blot analysis. A representative blot out of three independently performed experiments is shown.

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 8: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

554E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

Trk family receptor expression (Table 2). In addition, 1/65 (=1.5%) SI-NETs also stained positive with the pan-Trk antibody and 1/3 appendix NETs (=33.3%) (Table 2). Taken together, our results demonstrate that a total of 12.2% (13/107) of NET samples stained positive for receptors of the Trk family (Table 2).

Discussion

Here, we report on the potential role of Trk inhibitors as new molecular targeting approach for NETs of the GEP system. We evaluated the effects of the specific pan-Trk inhibitor GNF-5837 either alone or in a dual-targeting

approach on major cellular signaling mechanisms such as the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway as well as cell cycle components. Furthermore, we evaluated the prevalence of Trks immunohistochemically in a large patient cohort and could assess possible predictive markers for personalized molecular targeted therapy with Trk inhibitors.

In the first part of this study, effects of the highly selective pan-Trk inhibitor GNF-5837 (Albaugh et  al. 2012) were investigated in human neuroendocrine tumor cell lines in vitro.

GNF-5837 significantly decreased GOT1 cell survival (Fig.  1 and Table  1) due to TrkA inhibition (Fig.  2A).

Figure 5(A) Cell survival of GOT1 cells with single substance treatment of pan-Trk inhibitor GNF-5837 (500 nM) or everolimus (10 nM) and in combination (GNF-5837 + everolimus) after 144 h of incubation. The arithmetic means and s.d. of at least three independent experiments are shown. Statistical significant different results in comparison to either single substance treatment are shown, considering *P < 0.05; **P < 0.01; ***P < 0.001. (B) Effects of the combinational treatment (GNF-5837 + everolimus) on the expression level of different components from the cell cycle, the apoptotic response and the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway after 24 h of incubation in GOT1 cells. Basal protein expression level of proteins was evaluated by Western blot analysis. A representative blot out of three independently performed experiments is shown.

Figure 6Expression level of TrkA in GEP-NET tumor samples. (A) TrkA strong positive (score = 3) sample. (B) TrkA negative (score = 0) sample.

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 9: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

555E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

In contrast, lack of Trk expression in BON1 and NCI-727 cell lines (Fig. 2A) and thus obvious independence of cell growth from Trk family receptors caused GNF-5837 treatment resistance in BON1 and NCI-727 cell lines (Supplementary Fig.  1) and confirmed GNF-5837 to be a highly selective pan-Trk inhibitor (Albaugh et al. 2012). The anti-proliferative effects of Trk inhibition in neuroendocrine GOT1 tumor cells are in accordance with findings in other preclinical cancer cell models (Dolle et al. 2003, Albaugh et  al. 2012, Seo  et  al. 2013, Shargh et  al. 2016). Accordingly, Trk overexpression showed to enhance tumor growth, metastasis formation and invasiveness in different tumor entities in vitro (Sortino et al. 2000, Lagadec et al. 2009). In the GOT1 cells from a concentration of 50 nM on a plateau effect is observed, where further concentration augmentation did not yield a better cell killing effect (Fig. 1). This might be explained by a maximum TrkA receptor occupation of GNF-5837 at this concentration (Maliartchouk et al. 2000).

Incubation of GOT1 cells with GNF-5837 caused inhibition of TrkA activity as demonstrated by a decrease of TrkA phosphorylation status (Fig.  3A and C), while only specific NGF stimulation caused TrkA phosphorylation (Fig. 3A and B). In contrast, GNF-5837 caused a significant increase of total un-phosphorylated TrkA receptor expression levels in GOT1 cells, possibly

due to compensatory feedback mechanisms (Fig.  3C). Similar feedback loops with an upregulated re-expression of TrkA after inhibition have been reported in human airway smooth muscle cells (Freund-Michel & Frossard 2008). In Trk receptor-deficient BON1 and H727 cells, NGF showed a strong expression level (Fig. 2B). Here, we hypothesize that the high expression levels of NGF might be due to the lack of the specific receptor (TrkA), trying to compensate for the missing cellular TrkA-NGF signaling

Table 2 TrkA and pan-Trk expression level in different primary tumor localizations from 107 patient probes.

Primary tumor localization TotalSmall intestine Pancreas CUP Stomach Colon Appendix

TrkA expression level

TrkA negative n = 65 n = 25 n = 2 n = 1 n = 3 n = 3 n = 99 TrkA weak

expressionn = 0 n = 7 n = 0 n = 0 n = 0 n = 0 n = 7

TrkA moderate expression

n = 0 n = 1 n = 0 n = 0 n = 0 n = 0 n = 1

Total TrkA positive

n = 0/65 (=0%) n = 8/33 (=24.2%) n = 0/2 (=0%) n = 0/1 (=0%) n = 0/3 (=0%) n = 0/3 (=0%) n = 8/107 (=7.5%)

pan-Trk expression level

pan-Trk negative

n = 64 n = 22 n = 2 n = 1 n = 3 n = 2 n = 94

pan-Trk weak expression

n = 0 n = 9 n = 0 n = 0 n = 0 n = 0 n = 9

pan-Trk Moderate expression

n = 0 n = 1 n = 0 n = 0 n = 0 n = 1 n = 2

pan-Trk strong expression

n = 1 n = 1 n = 0 n = 0 n = 0 n = 0 n = 2

Total pan-Trk positive

n = 1/65 (=1.5%)

n = 11/33 (=33.3%)

n = 0/2 (=0%) n = 0/1 (=0%) n = 0/3 (=0%) n = 1/3 (=33.3%)

n = 13/107 (=12.2%)

Cancer of unknown primary tumor are designated ‘CUP’.

Figure 7Cell cycle analysis measured by flow cytometry. The arithmetic means and s.d. of at least three independent experiments are shown. Statistical significant different results in comparison to the Control DMSO are shown, considering *P < 0.05; **P < 0.01; ***P < 0.001. A full color version of this figure is available at https://doi.org/10.1530/ERC-17-0201.

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 10: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

556E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

cascade. Tagliabue et al. found similar results when breast cancer cells with poor TrkA expression showed high NGF expression in order to recruit other receptors to induce cancer cell growth (Tagliabue et al. 2000).

The PI3K-Akt-mTOR and the Ras-Raf-MEK-ERK pathway are often deregulated in human cancers including NETs, and the importance of these pathways comprises cellular oncogenic modulation mechanisms such as cell growth, survival, proliferation, angiogenesis and motility (Cantley 2002, Van Gompel  et  al. 2005, Kunnimalaiyaan  et  al. 2007, Zitzmann et  al. 2007, 2011, Cook  et  al. 2010, Iida  et  al. 2012, Valentino  et  al. 2014, Briest & Grabowski 2014). TrkA activation and downstream signaling upon NGF stimulation has been shown in multiple cell types to mediate its proliferative and anti-apoptotic characteristics mainly through PI3K-Akt-mTOR and Ras-Raf-MEK-ERK cascade signaling (Sofroniew  et  al. 2001, Huang & Reichardt 2003, Rahbek  et  al. 2005, Jang  et  al. 2007, Lawrence  et  al. 2008, Julio-Pieper  et  al. 2009, Amatu et  al. 2016, Demir et  al. 2016, Pediaditakis  et  al. 2016). In accordance to these data, in GOT1 cells, NGF stimulation induced the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK signaling cascade (Fig.  4B), while inhibition of TrkA signaling by the pan-Trk inhibitor GNF-5837 caused downregulation of both downstream signaling pathways (Fig.  4A). Accordingly, in a colorectal cancer cell line (KM12) and a LCNEC cell line, pan-Trk inhibition with AZD7451 leads to a cellular survival decrease through pErk or pAkt downregulation (Tatematsu et al. 2014). As a consequence to PI3K-Akt-mTOR and Ras-Raf-MEK-ERK alteration, downstream cell cycle component Cyclin D3 was upregulated upon NGF stimulation or downregulated upon GNF-5837 inhibition, respectively (Fig.  4). Furthermore, a significant population increase in the cell cycle phase G1 confirms a cell cycle downregulation upon GNF-5837 treatment (Fig. 7). GOT1 cells showed not only a strong TrkA expression but also a slight expression of TrkC (Fig. 2A). Besides inhibition of TrkA, also inhibition of TrkC, might contribute to Ras-Raf-MEK-ERK and PI3K-Akt-mTOR pathway inhibition following incubation with the pan-Trk inhibitor GNF-5837. In adenoid cystic carcinoma cells, TrkC signaling was activated and required its ligand NT-3 to stimulate invasive behavior through Ras-Raf-MEK-ERK and PI3K-Akt-mTOR signaling (Ivanov et al. 2013).

Interestingly, the cyclin-dependent kinase inhibitor p21 is upregulated upon NGF stimulation (Fig.  4B and Supplementary Fig.  3) and downregulated upon Trk inhibition by GNF-5837 treatment (Fig.  4A and

Supplementary Fig.  2). This fact might be explained by induction of the anti-apoptotic Bcl2 after NGF stimulation and the downregulation of Bcl2 and augmentation of the Sub-G1 population after GNF-5837 inhibition (Figs 4 and 7). Studies have shown that in some systems, the induction of apoptotic mechanisms relate with p21 inhibition, counteracting its tumor-suppressive function (Gartel & Tyner 2002).

Due to the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation upon Trk inhibition by GNF-5837 alone (Fig.  4A and Supplementary Fig.  2), we aimed to further investigate the effects of GNF-5837 in a dual-targeting approach with the mTORC1 inhibitor everolimus. The mTOR inhibitor everolimus has proven anti-tumoral efficacy in several clinical phase 3 trials in patients with NET (Pavel et al. 2011, Yao  et  al. 2016a,b). In NET cell lines, everolimus has been shown to inhibit mTOR downstream signaling and to cause a compensatory upregulation of the Ras-Raf-MEK-ERK signaling cascade (Zitzmann  et  al. 2010, Wagle  et  al. 2014, Capozzi  et  al. 2015, He  et  al. 2016, Vandamme et al. 2016, Zatelli et al. 2016). Furthermore, development of resistance against everolimus during long-term treatment is often observed in NETs and possibly caused by feedback loops on the PI3K-Akt axis (Zitzmann et al. 2010, Wagle et al. 2014, Capozzi et al. 2015, He et al. 2016, Vandamme et al. 2016, Zatelli et al. 2016). Thus, the potential benefit of a dual-targeting approach is not only to enhance single substance treatment efficacy, but also to prevent and/or counteract resistance mechanisms. We found that the combinational approach of the Trk inhibitor GNF-5837 and the mTOR inhibitor everolimus caused additive anti-proliferative effects (Fig.  5) and agonistically downregulated the PI3K-Akt-mTOR pathway, as well as Cyclin D3 (Fig. 5B and Supplementary Fig. 4). In a Trk-induced leukemia mouse-model, the tumor cells showed a high susceptibility toward the mTORC1 inhibitor rapamycin and RAD001 (everolimus) via PI3K-Akt-mTOR pathway and Ras-Raf-MEK-ERK downregulation (Rhein et al. 2011).

In the second part of this study, we examined the expression of Trk family receptors in a tissue microarray (TMA) encompassing 107 tumor probes of patients with GEP-NETs.

Using a specific TrkA antibody, a positive staining in 8/107 tumor probes was exclusively due to the sub-group of pNETs with a positive staining in 8/33 (24%) of all pNETs (Table 2). Using a pan-Trk antibody (detecting endogenous levels of total Trk protein encompassing TrkA, TrkB and TrkC) a positive staining in 13/107 tumor probes

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 11: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

557E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

was mainly due to the subset of pNETs with a positive staining in 11/33 (33%) of all pNETs (Table 2). These data demonstrate a high prevalence of Trk receptor expression in pancreatic NETs. Therefore, immunohistochemical or molecular screening for Trk expression in pNETs might be useful as a predictive marker to select patients with pNETs expressing Trks for molecular targeted therapy with Trk inhibitors. Whether Trk overexpression or overactivation caused by chromosomal rearrangements and NTRK gene fusions (Amatu et al. 2016) is also existent in NET should be investigated in further studies. Interestingly, in the malignant neuroendocrine Merkel cell carcinoma, expression of TrkA was found in all tested tumor specimens (36/36), underlining a possible therapeutic relevance of Trk blockage in NETs not only from the GEP system (Wehkamp et al. 2017).

A limitation of our study is the fact that currently available human neuroendocrine tumor cell lines might not reflect the actual biological expression state of Trk receptors in neuroendocrine tumors in vivo. While the SI carcinoid cell line GOT1 was the only tested cell line expressing a functional Trk receptor (Figs 1, 2A and 3), the TMA analysis revealed Trk expression in SI NETs to be a very rare event as only 1/65 (1.5%) of SI NET tumor probes showed a positive staining with the pan-Trk antibody (Table  2). Further investigation regarding the effects of GNF-5837 in NETs is required in order to validate molecular target credentials and clinical relevance.

Taken together our findings, the highly selective pan-Trk inhibitor GNF-5837 presents significant anti-proliferative efficacy in human neuroendocrine GOT1 cells in vitro. Inhibition of TrkA signaling in GOT1 cells caused subsequent downregulation of the PI3K-Akt-mTOR and Ras-Raf-MEK-ERK signaling cascades, as well as the cell cycle and upregulation of apoptosis. A dual-targeting approach using the pan-Trk inhibitor GNF-5837 plus the mTOR inhibitor everolimus showed additive inhibitory effects on PI3K-Akt-mTOR pathway and cyclin D3. Tissue microarray analysis revealed Trk expression in 24% (TrkA)/33% (pan-Trk) of pancreatic NETs. Immunohistochemical or molecular screening for Trk expression in pNETs might be useful as a predictive marker to select patients with pNETs expressing Trks for molecular targeted therapy with Trk inhibitors.

Supplementary dataThis is linked to the online version of the paper at https://doi.org/10.1530/ERC-17-0201.

Declaration of interestC J A has received research contracts (Ipsen, Novartis), lecture honorarium (Ipsen, Novartis, Pfizer, Falk) and advisory board honorarium (Novartis). The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work.

FundingThis study has been funded by the Theranostic Research Network, Bad Berka, Germany – Wilhelm und Ingeburg Dinse Gedächtnis-Stiftung. The biosampling of this study has been initially funded within NeoExNET (Network of Excellence for Neuroendocrine Tumors in Munich). NeoExNET is a national database for the evaluation of diagnostics, treatment and outcome in neuroendocrine tumors. NeoExNET is supported by the German Federal Ministry of Education and Research (BMBF; m4 Cluster – Personalized Medicine and Targeted Therapies Leading-Edge Cluster Munich, 16EX1221J, 01EX1020E) and by an unrestricted educational grant of Novartis Pharma GmbH, Nürnberg, Germany. Members of the NeoExNET-Study group include: Principal investigator: Günter K Stalla (Max Planck Institute of Psychiatry, Munich). Steering Committee: Felix Beuschlein (Ludwig Maximilian University, Munich), Christoph Auernhammer (Ludwig Maximilian University, Munich), Klaus A Kuhn (Technische Universität München, Munich).

ReferencesAlbaugh P, Fan Y, Mi Y, Sun F, Adrian F, Li N, Jia Y, Sarkisova Y,

Kreusch A, Hood T, et al. 2012 Discovery of GNF-5837, a selective TRK inhibitor with efficacy in rodent cancer tumor models. ACS Medicinal Chemistry Letters 3 140–145. (https://doi.org/10.1021/ml200261d)

Amatu A, Sartore-Bianchi A & Siena S 2016 NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 1 e000023. (https://doi.org/10.1136/esmoopen-2015-000023)

Ardini E, Menichincheri M, Banfi P, Bosotti R, De Ponti C, Pulci R, Ballinari D, Ciomei M, Texido G, Degrassi A, et al. 2016 Entrectinib, a Pan-TRK, ROS1, and ALK inhibitor with activity in multiple molecularly defined cancer indications. Molecular Cancer Therapeutics 15 628–639. (https://doi.org/10.1158/1535-7163.MCT-15-0758)

Auernhammer CJ & Goke B 2011 Therapeutic strategies for advanced neuroendocrine carcinomas of jejunum/ileum and pancreatic origin. Gut 60 1009–1021. (https://doi.org/10.1136/gut.2009.204453)

Barbacid M 1994 The Trk family of neurotrophin receptors. Journal of Neurobiology 25 1386–1403. (https://doi.org/10.1002/neu.480251107)

Bibel M & Barde YA 2000 Neurotrophins: key regulators of cell fate and cell shape in the vertebrate nervous system. Genes and Development 14 2919–2937. (https://doi.org/10.1101/gad.841400)

Bodei L, Cremonesi M, Kidd M, Grana CM, Severi S, Modlin IM & Paganelli G 2014 Peptide receptor radionuclide therapy for advanced neuroendocrine tumors. Thoracic Surgery Clinics 24 333–349. (https://doi.org/10.1016/j.thorsurg.2014.04.005)

Briest F & Grabowski P 2014 PI3K-AKT-mTOR-signaling and beyond: the complex network in gastroenteropancreatic neuroendocrine neoplasms. Theranostics 4 336–365. (https://doi.org/10.7150/thno.7851)

Cakir M & Grossman A 2011 The molecular pathogenesis and management of bronchial carcinoids. Expert Opinion on Therapeutic Targets 15 457–491. (https://doi.org/10.1517/14728222.2011.555403)

Cantley LC 2002 The phosphoinositide 3-kinase pathway. Science 296 1655–1657. (https://doi.org/10.1126/science.296.5573.1655)

Capozzi M, Caterina I, De Divitiis C, von Arx C, Maiolino P, Tatangelo F, Cavalcanti E, Di Girolamo E, Iaffaioli RV, Scala S, et al. 2015 Everolimus and pancreatic neuroendocrine tumors (PNETs): activity,

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 12: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

558E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

resistance and how to overcome it. International Journal of Surgery 21 (Supplement 1) S89–S94. (https://doi.org/10.1016/j.ijsu.2015.06.064)

Cives M & Strosberg J 2017 Radionuclide therapy for neuroendocrine tumors. Current Oncology Reports 19 9. (https://doi.org/10.1007/s11912-017-0567-8)

Clary DO & Reichardt LF 1994 An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3. PNAS 91 11133–11137. (https://doi.org/10.1073/pnas.91.23.11133)

Cook MR, Pinchot SN, Jaskula-Sztul R, Luo J, Kunnimalaiyaan M & Chen H 2010 Identification of a novel Raf-1 pathway activator that inhibits gastrointestinal carcinoid cell growth. Molecular Cancer Therapeutics 9 429–437. (https://doi.org/10.1158/1535-7163.MCT-09-0718)

Davidson B, Lazarovici P, Ezersky A, Nesland JM, Berner A, Risberg B, Trope CG, Kristensen GB, Goscinski M, van de Putte G, et al. 2001 Expression levels of the nerve growth factor receptors TrkA and p75 in effusions and solid tumors of serous ovarian carcinoma patients. Clinical Cancer Research 7 3457–3464.

Demir IE, Tieftrunk E, Schorn S, Friess H & Ceyhan GO 2016 Nerve growth factor & TrkA as novel therapeutic targets in cancer. Biochimica et Biophysica Acta 1866 37–50. (https://doi.org/10.1016/j.bbcan.2016.05.003)

Dolle L, El Yazidi-Belkoura I, Adriaenssens E, Nurcombe V & Hondermarck H 2003 Nerve growth factor overexpression and autocrine loop in breast cancer cells. Oncogene 22 5592–5601. (https://doi.org/10.1038/sj.onc.1206805)

Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, Bauer TM, Farago AF, Wheler JJ, Liu SV, et al. 2017 Safety and antitumor activity of the multi-targeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib (RXDX-101): combined results from two phase 1 trials (ALKA-372-001 and STARTRK-1). Cancer Discovery 7 400–409. (https://doi.org/10.1158/2159-8290.CD-16-1237)

Evers BM, Townsend CM Jr, Upp JR, Allen E, Hurlbut SC, Kim SW, Rajaraman S, Singh P, Reubi JC & Thompson JC 1991 Establishment and characterization of a human carcinoid in nude mice and effect of various agents on tumor growth. Gastroenterology 101 303–311. (https://doi.org/10.1016/0016-5085(91)90004-5)

Freund-Michel V & Frossard N 2008 Overexpression of functional TrkA receptors after internalisation in human airway smooth muscle cells. Biochimica et Biophysica Acta 1783 1964–1971. (https://doi.org/10.1016/j.bbamcr.2008.05.014)

Gartel AL & Tyner AL 2002 The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Molecular Cancer Therapeutics 1 639–649.

Grozinsky-Glasberg S, Shimon I & Rubinfeld H 2012 The role of cell lines in the study of neuroendocrine tumors. Neuroendocrinology 96 173–187. (https://doi.org/10.1159/000338793)

He K, Chen D, Ruan H, Li X, Tong J, Xu X, Zhang L & Yu J 2016 BRAFV600E-dependent Mcl-1 stabilization leads to everolimus resistance in colon cancer cells. Oncotarget 7 47699–47710. (https://doi.org/10.18632/oncotarget.10277)

Huang EJ & Reichardt LF 2003 Trk receptors: roles in neuronal signal transduction. Annual Review of Biochemistry 72 609–642. (https://doi.org/10.1146/annurev.biochem.72.121801.161629)

Iida S, Miki Y, Ono K, Akahira J, Nakamura Y, Suzuki T & Sasano H 2012 Synergistic anti-tumor effects of RAD001 with MEK inhibitors in neuroendocrine tumors: a potential mechanism of therapeutic limitation of mTOR inhibitor. Molecular and Cellular Endocrinology 350 99–106. (https://doi.org/10.1016/j.mce.2011.11.024)

Ivanov SV, Panaccione A, Brown B, Guo Y, Moskaluk CA, Wick MJ, Brown JL, Ivanova AV, Issaeva N, El-Naggar AK, et al. 2013 TrkC signaling is activated in adenoid cystic carcinoma and requires NT-3 to stimulate invasive behavior. Oncogene 32 3698–3710. (https://doi.org/10.1038/onc.2012.377)

Jang SW, Okada M, Sayeed I, Xiao G, Stein D, Jin P & Ye K 2007 Gambogic amide, a selective agonist for TrkA receptor that possesses

robust neurotrophic activity, prevents neuronal cell death. PNAS 104 16329–16334. (https://doi.org/10.1073/pnas.0706662104)

Julio-Pieper M, Lozada P, Tapia V, Vega M, Miranda C, Vantman D, Ojeda SR & Romero C 2009 Nerve growth factor induces vascular endothelial growth factor expression in granulosa cells via a trkA receptor/mitogen-activated protein kinase-extracellularly regulated kinase 2-dependent pathway. Journal of Clinical Endocrinology and Metabolism 94 3065–3071. (https://doi.org/10.1210/jc.2009-0542)

Kaplan DR, Hempstead BL, Martin-Zanca D, Chao MV & Parada LF 1991 The trk proto-oncogene product: a signal transducing receptor for nerve growth factor. Science 252 554–558. (https://doi.org/10.1126/science.1850549)

Karpathakis A, Dibra H, Pipinikas C, Feber A, Morris T, Francis J, Oukrif D, Mandair D, Pericleous M, Mohmaduvesh M, et al. 2017 Progressive epigenetic dysregulation in neuroendocrine tumour liver metastases. Endocrine-Related Cancer 24 L21–L25. (https://doi.org/10.1530/ERC-16-0419)

Khotskaya YB, Holla VR, Farago AF, Mills Shaw KR, Meric-Bernstam F & Hong DS 2017 Targeting TRK family proteins in cancer. Pharmacology and Therapeutics 173 58–66. (https://doi.org/10.1016/j.pharmthera.2017.02.006)

Kidd M, Modlin I & Oberg K 2016 Towards a new classification of gastroenteropancreatic neuroendocrine neoplasms. Nature Reviews Clinical Oncology 13 691–705. (https://doi.org/10.1038/nrclinonc.2016.85)

Klein R, Jing SQ, Nanduri V, O’Rourke E & Barbacid M 1991 The trk proto-oncogene encodes a receptor for nerve growth factor. Cell 65 189–197. (https://doi.org/10.1016/0092-8674(91)90419-Y)

Knosel T, Emde A, Schluns K, Chen Y, Jurchott K, Krause M, Dietel M & Petersen I 2005 Immunoprofiles of 11 biomarkers using tissue microarrays identify prognostic subgroups in colorectal cancer. Neoplasia 7 741–747. (https://doi.org/10.1593/neo.05178)

Knosel T, Emde V, Schluns K, Schlag PM, Dietel M & Petersen I 2006 Cytokeratin profiles identify diagnostic signatures in colorectal cancer using multiplex analysis of tissue microarrays. Cellular Oncology 28 167–175. (https://doi.org/10.1155/2006/354295)

Kolby L, Bernhardt P, Ahlman H, Wangberg B, Johanson V, Wigander A, Forssell-Aronsson E, Karlsson S, Ahren B, Stenman G, et al. 2001 A transplantable human carcinoid as model for somatostatin receptor-mediated and amine transporter-mediated radionuclide uptake. American Journal of Pathology 158 745–755. (https://doi.org/10.1016/S0002-9440(10)64017-5)

Kunnimalaiyaan M, Ndiaye M & Chen H 2007 Neuroendocrine tumor cell growth inhibition by ZM336372 through alterations in multiple signaling pathways. Surgery 142 959–964. (https://doi.org/10.1016/j.surg.2007.09.020)

Lagadec C, Meignan S, Adriaenssens E, Foveau B, Vanhecke E, Romon R, Toillon RA, Oxombre B, Hondermarck H & Le Bourhis X 2009 TrkA overexpression enhances growth and metastasis of breast cancer cells. Oncogene 28 1960–1970. (https://doi.org/10.1038/onc.2009.61)

Lawrence MC, McGlynn K, Shao C, Duan L, Naziruddin B, Levy MF & Cobb MH 2008 Chromatin-bound mitogen-activated protein kinases transmit dynamic signals in transcription complexes in beta-cells. PNAS 105 13315–13320. (https://doi.org/10.1073/pnas.0806465105)

Maliartchouk S, Debeir T, Beglova N, Cuello AC, Gehring K & Saragovi HU 2000 Genuine monovalent ligands of TrkA nerve growth factor receptors reveal a novel pharmacological mechanism of action. Journal of Biological Chemistry 275 9946–9956. (https://doi.org/10.1074/jbc.275.14.9946)

Martin-Zanca D, Oskam R, Mitra G, Copeland T & Barbacid M 1989 Molecular and biochemical characterization of the human trk proto-oncogene. Molecular and Cellular Biology 9 24–33. (https://doi.org/10.1128/MCB.9.1.24)

McGregor LM, McCune BK, Graff JR, McDowell PR, Romans KE, Yancopoulos GD, Ball DW, Baylin SB & Nelkin BD 1999 Roles of trk family neurotrophin receptors in medullary thyroid carcinoma

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 13: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

559E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

development and progression. PNAS 96 4540–4545. (https://doi.org/10.1073/pnas.96.8.4540)

Modlin IM, Moss SF, Chung DC, Jensen RT & Snyderwine E 2008a Priorities for improving the management of gastroenteropancreatic neuroendocrine tumors. Journal of the National Cancer Institute 100 1282–1289. (https://doi.org/10.1093/jnci/djn275)

Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, Caplin M, Delle Fave G, Kaltsas GA, Krenning EP, et al. 2008b Gastroenteropancreatic neuroendocrine tumours. Lancet Oncology 9 61–72. (https://doi.org/10.1016/S1470-2045(07)70410-2)

Numakawa T, Suzuki S, Kumamaru E, Adachi N, Richards M & Kunugi H 2010 BDNF function and intracellular signaling in neurons. Histology and Histopathology 25 237–258. (https://doi.org/10.14670/HH-25.237)

Odate S, Nakamura K, Onishi H, Kojima M, Uchiyama A, Nakano K, Kato M, Tanaka M & Katano M 2013 TrkB/BDNF signaling pathway is a potential therapeutic target for pulmonary large cell neuroendocrine carcinoma. Lung Cancer 79 205–214. (https://doi.org/10.1016/j.lungcan.2012.12.004)

Oelmann E, Sreter L, Schuller I, Serve H, Koenigsmann M, Wiedenmann B, Oberberg D, Reufi B, Thiel E & Berdel WE 1995 Nerve growth factor stimulates clonal growth of human lung cancer cell lines and a human glioblastoma cell line expressing high-affinity nerve growth factor binding sites involving tyrosine kinase signaling. Cancer Research 55 2212–2219.

Osborne JK, Larsen JE, Shields MD, Gonzales JX, Shames DS, Sato M, Kulkarni A, Wistuba II, Girard L, Minna JD, et al. 2013 NeuroD1 regulates survival and migration of neuroendocrine lung carcinomas via signaling molecules TrkB and NCAM. PNAS 110 6524–6529. (https://doi.org/10.1073/pnas.1303932110)

Passiglia F, Caparica R, Giovannetti E, Giallombardo M, Listi A, Diana P, Cirrincione G, Caglevic C, Raez LE, Russo A, et al. 2016 The potential of neurotrophic tyrosine kinase (NTRK) inhibitors for treating lung cancer. Expert Opinion on Investigational Drugs 25 385–392. (https://doi.org/10.1517/13543784.2016.1152261)

Pavel ME & Sers C 2016 WOMEN IN CANCER THEMATIC REVIEW: systemic therapies in neuroendocrine tumors and novel approaches toward personalized medicine. Endocrine-Related Cancer 23 T135–T154. (https://doi.org/10.1530/ERC-16-0370)

Pavel ME, Hainsworth JD, Baudin E, Peeters M, Horsch D, Winkler RE, Klimovsky J, Lebwohl D, Jehl V, Wolin EM, et al. 2011 Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378 2005–2012. (https://doi.org/10.1016/S0140-6736(11)61742-X)

Pediaditakis I, Efstathopoulos P, Prousis KC, Zervou M, Arevalo JC, Alexaki VI, Nikoletopoulou V, Karagianni E, Potamitis C, Tavernarakis N, et al. 2016 Selective and differential interactions of BNN27, a novel C17-spiroepoxy steroid derivative, with TrkA receptors, regulating neuronal survival and differentiation. Neuropharmacology 111 266–282. (https://doi.org/10.1016/j.neuropharm.2016.09.007)

Rahbek UL, Dissing S, Thomassen C, Hansen AJ & Tritsaris K 2005 Nerve growth factor activates aorta endothelial cells causing PI3K/Akt- and ERK-dependent migration. Pflügers Archiv 450 355–361. (https://doi.org/10.1007/s00424-005-1436-0)

Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, et al. 2011 Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. New England Journal of Medicine 364 501–513. (https://doi.org/10.1056/NEJMoa1003825)

Rhein M, Schwarzer A, Yang M, Kaever V, Brugman M, Meyer J, Ganser A, Baum C & Li Z 2011 Leukemias induced by altered TRK-signaling are sensitive to mTOR inhibitors in preclinical models. Annals of Hematology 90 283–292. (https://doi.org/10.1007/s00277-010-1065-3)

Schimmack S, Svejda B, Lawrence B, Kidd M & Modlin IM 2011 The diversity and commonalities of gastroenteropancreatic

neuroendocrine tumors. Langenbeck’s Archives of Surgery 396 273–298. (https://doi.org/10.1007/s00423-011-0739-1)

Seo JH, Jung KH, Son MK, Yan HH, Ryu YL, Kim J, Lee JK, Hong S & Hong SS 2013 Anti-cancer effect of HS-345, a new tropomyosin-related kinase A inhibitor, on human pancreatic cancer. Cancer Letters 338 271–281. (https://doi.org/10.1016/j.canlet.2013.04.002)

Shargh VH, Hondermarck H & Liang M 2016 Albumin hybrid nanoparticles loaded with tyrosine kinase A inhibitor GNF-5837 for targeted inhibition of breast cancer cell growth and invasion. International Journal of Pharmaceutics 515 527–534. (https://doi.org/10.1016/j.ijpharm.2016.10.057)

Sofroniew MV, Howe CL & Mobley WC 2001 Nerve growth factor signaling, neuroprotection, and neural repair. Annual Review of Neuroscience 24 1217–1281. (https://doi.org/10.1146/annurev.neuro.24.1.1217)

Sortino MA, Condorelli F, Vancheri C, Chiarenza A, Bernardini R, Consoli U & Canonico PL 2000 Mitogenic effect of nerve growth factor (NGF) in LNCaP prostate adenocarcinoma cells: role of the high- and low-affinity NGF receptors. Molecular Endocrinology 14 124–136. (https://doi.org/10.1210/mend.14.1.0402)

Spampatti M, Vlotides G, Spottl G, Maurer J, Goke B & Auernhammer CJ 2014 Aspirin inhibits cell viability and mTOR downstream signaling in gastroenteropancreatic and bronchopulmonary neuroendocrine tumor cells. World Journal of Gastroenterology 20 10038–10049. (https://doi.org/10.3748/wjg.v20.i29.10038)

Tagliabue E, Castiglioni F, Ghirelli C, Modugno M, Asnaghi L, Somenzi G, Melani C & Menard S 2000 Nerve growth factor cooperates with p185(HER2) in activating growth of human breast carcinoma cells. Journal of Biological Chemistry 275 5388–5394. (https://doi.org/10.1074/jbc.275.8.5388)

Tatematsu T, Sasaki H, Shimizu S, Okuda K, Shitara M, Hikosaka Y, Moriyama S, Yano M, Brown J & Fujii Y 2014 Investigation of neurotrophic tyrosine kinase receptor 1 fusions and neurotrophic tyrosine kinase receptor family expression in non-small-cell lung cancer and sensitivity to AZD7451 in vitro. Molecular and Clinical Oncology 2 725–730. (https://doi.org/10.3892/mco.2014.318)

Valentino JD, Li J, Zaytseva YY, Mustain WC, Elliott VA, Kim JT, Harris JW, Campbell K, Weiss H, Wang C, et al. 2014 Cotargeting the PI3K and RAS pathways for the treatment of neuroendocrine tumors. Clinical Cancer Research 20 1212–1222. (https://doi.org/10.1158/1078-0432.CCR-13-1897)

Van Gompel JJ, Kunnimalaiyaan M, Holen K & Chen H 2005 ZM336372, a Raf-1 activator, suppresses growth and neuroendocrine hormone levels in carcinoid tumor cells. Molecular Cancer Therapeutics 4 910–917. (https://doi.org/10.1158/1535-7163.MCT-04-0334)

van Vliet EI, Teunissen JJ, Kam BL, de Jong M, Krenning EP & Kwekkeboom DJ 2013 Treatment of gastroenteropancreatic neuroendocrine tumors with peptide receptor radionuclide therapy. Neuroendocrinology 97 74–85. (https://doi.org/10.1159/000335018)

Vandamme T, Beyens M, de Beeck KO, Dogan F, van Koetsveld PM, Pauwels P, Mortier G, Vangestel C, de Herder W, Van Camp G, et al. 2016 Long-term acquired everolimus resistance in pancreatic neuroendocrine tumours can be overcome with novel PI3K-AKT-mTOR inhibitors. British Journal of Cancer 114 650–658. (https://doi.org/10.1038/bjc.2016.25)

Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A, Rosenberg M, Gray N, Barletta JA, Guo Y, Swanson SJ, et al. 2014 Response and acquired resistance to everolimus in anaplastic thyroid cancer. New England Journal of Medicine 371 1426–1433. (https://doi.org/10.1056/NEJMoa1403352)

Wehkamp U, Stern S, Kruger S, Hauschild A, Rocken C & Egberts F 2017 Tropomyosin receptor kinase A expression on merkel cell carcinoma cells. JAMA Dermatology 153 1166–1169. (https://doi.org/10.1001/jamadermatol.2017.2495)

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access

Page 14: Tropomyosin receptor kinase: a novel target in screened … · V Heinzle et al Tropomyosin receptor kinase (Trk) in NETs Endocrine-Related 25:5 Cancer Introduction Neuroendocrine

Printed in Great BritainPublished by Bioscientifica Ltd.https://doi.org/10.1530/ERC-17-0201

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology

560E T Aristizabal Prada, V Heinzle et al.

Tropomyosin receptor kinase (Trk) in NETs

25:5Endocrine-Related Cancer

Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, Abdalla EK, Fleming JB, Vauthey JN, Rashid A, et al. 2008 One hundred years after ‘carcinoid’: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. Journal of Clinical Oncology 26 3063–3072. (https://doi.org/10.1200/JCO.2007.15.4377)

Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, et al. 2011 Everolimus for advanced pancreatic neuroendocrine tumors. New England Journal of Medicine 364 514–523. (https://doi.org/10.1056/NEJMoa1009290)

Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi C, Wolin E, Tomasek J, Raderer M, Lahner H, Voi M, et al. 2016a Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 387 968–977. (https://doi.org/10.1016/S0140-6736(15)00817-X)

Yao JC, Pavel M, Lombard-Bohas C, Van Cutsem E, Voi M, Brandt U, He W, Chen D, Capdevila J, de Vries EG, et al. 2016b Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 Study. Journal of Clinical Oncology 34 3609–3913. (https://doi.org/10.1200/JCO.2016.68.0702)

Zatelli MC, Fanciulli G, Malandrino P, Ramundo V, Faggiano A & Colao A 2016 Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocrine-Related Cancer 23 R173–R183. (https://doi.org/10.1530/ERC-15-0413)

Zhu ZW, Friess H, Wang L, Di Mola FF, Zimmermann A & Buchler MW 2000 Down-regulation of nerve growth factor in poorly differentiated and advanced human esophageal cancer. Anticancer Research 20 125–132.

Zitzmann K, De Toni EN, Brand S, Goke B, Meinecke J, Spottl G, Meyer HH & Auernhammer CJ 2007 The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology 85 54–60. (https://doi.org/10.1159/000100057)

Zitzmann K, Ruden J, Brand S, Goke B, Lichtl J, Spottl G & Auernhammer CJ 2010 Compensatory activation of Akt in response to mTOR and Raf inhibitors – a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Letters 295 100–109. (https://doi.org/10.1016/j.canlet.2010.02.018)

Zitzmann K, de Toni E, von Ruden J, Brand S, Goke B, Laubender RP & Auernhammer CJ 2011 The novel Raf inhibitor Raf265 decreases Bcl-2 levels and confers TRAIL-sensitivity to neuroendocrine tumour cells. Endocrine-Related Cancer 18 277–285. (https://doi.org/10.1530/ERC-10-0108)

Received in final form 9 March 2018Accepted 21 March 2018Accepted Preprint published online 21 March 2018

Downloaded from Bioscientifica.com at 08/02/2021 09:39:33AMvia free access