Cancer Letters

Cancer Letters

Volume 432, 28 September 2018, Pages 227-236
Cancer Letters

Original Articles
Targeting the NRG1/HER3 pathway in tumor cells and cancer-associated fibroblasts with an anti-neuregulin 1 antibody inhibits tumor growth in pre-clinical models of pancreatic cancer

https://doi.org/10.1016/j.canlet.2018.06.023Get rights and content

Highlights

  • NRG1 is expressed in human pancreatic tumors and cancer-associated fibroblasts.

  • NRG1 promotes pancreatic cell growth.

  • NRG1 trapping by 7E3 antibody inhibits HER3 activation and downstream signaling.

  • Targeting NRG1 inhibits growth of orthotopic pancreatic tumor xenografts.

Abstract

Neuregulin 1 (NRG1), a ligand for HER3 and HER4 receptors, is secreted by both pancreatic tumor cells (PC) and cancer-associated fibroblasts (CAFs), the latter representing the most abundant compound of pancreatic stroma. This desmoplastic stroma contributes to Pancreatic Ductal Adenocarcinoma (PDAC) aggressiveness and therapeutic failure by promoting tumor progression, invasion and resistance to chemotherapies. In the present work, we aimed at disrupting the complex crosstalk between PC and CAF in order to prevent tumor cell proliferation. To do so, we demonstrated the promising tumor growth inhibitory effect of the 7E3, an original antibody directed to NRG1. This antibody promotes antibody dependent cellular cytotoxicity in NRG1-positive PC and CAFs and inhibits NRG1-associated signaling pathway induction, by blocking NRG1-mediated HER3 activation. Moreover, 7E3 inhibits migration and growth of pancreatic cancer cells co-cultured with CAFs, both in vitro and in vivo using orthotopic pancreatic tumor xenografts. Our preclinical results demonstrate that the anti-NRG1 antibody 7E3 could represent a promising approach to target pancreatic stroma and cancer cells, thereby providing novel therapeutic options for PDAC.

Introduction

Pancreatic cancer remains one of the most aggressive tumors with an extremely poor prognosis and is estimated to become the second leading cause of cancer-related death by 2030 [1,2]. This dramatic outcome is related to the lack of efficient therapeutic tools, early diagnostic markers, and high resistance to chemotherapy.

Abundant desmoplastic reaction is a prominent feature of pancreatic cancer and constitutes up to 80% of the total tumor volume. It is characterized by the presence mainly of cancer-associated fibroblasts (CAFs), but also of immune cells, vasculature, and extracellular matrix (ECM). These stroma characteristics contribute to pancreatic cancer aggressivity and therapeutic failure because they promote tumor progression, invasion and resistance to chemotherapies [3,4]. Once activated, CAFs express alpha smooth muscle actin (αSMA) and secrete ECM and soluble factors (e.g., growth and inflammatory factors) that stimulate tumor growth and mediate inflammation. In preclinical studies, stroma disruption, by direct CAF targeting (hedgehog signaling inhibitors) [5] or by ECM enzymatic digestion (hyaluronidase), results in tumor inhibition and increases drug delivery [6]; however, controversial or disappointing results were reported by clinical trials. Moreover, recent studies have questioned CAF role in pancreatic tumor maintenance and highlighted the need of caution when targeting CAFs [[7], [8], [9]]. Indeed, several evidences indicate that there are different CAF subtypes with different functions [10,11]. Therefore, characterization of these subtypes is needed to enable the specific targeting of CAFs with pro-tumor features.

Neuregulins (NRGs) are large polypeptide growth factors that are part of the EGF family of proteins. They are encoded by four genes (NRG1-4) that are subject to extensive alternative mRNA splicing, leading to more than 30 variants grouped in six types (I-VI) with a distinct N-terminal part. NRG1 and 2 are both HER3 and HER4 ligands and have at least two splice alternatives in their EGF-like domains, resulting in α and β isoforms. NRG1 β has higher binding affinity for HER3 through HER2-dependent recruitment [12]. NRG3 and 4 bind only to HER4. Most isoforms are synthetized as transmembrane molecules with the EGF-like domain in the extracellular space and are released as soluble factors by cell surface proteases. Binding to the EGF-like domain of NRG1 promotes HER3 or HER4 conformational changes, leading to their dimerization and activation that result in the phosphorylation of downstream signaling molecules. Several evidences indicate that NRG1 contributes to the development and progression of different tumor types, such as breast, prostate or pancreatic cancer [[13], [14], [15], [16]]. High NRG1 expression has been correlated with poor prognosis in breast cancer, head and neck squamous cell carcinoma and pancreatic cancer [[13], [14], [15]]. In addition, NRG1 downregulation by antisense oligonucleotides, or NRG1 signaling inhibition by competitive binding of a shorter HER3 isoform reduces breast cancer cell proliferation and migration [16,17]. Furthermore, the crosstalk between NRG1 and HER3 is involved in resistance to chemo- and targeted therapies through the maintenance of oncogenic signaling pathways [18].

Several studies reported that not only EGF family growth factors but also their receptors play an important role in pancreatic cancer development. Expression of EGFR, HER2 and HER3 has been examined in pancreatic ductal carcinoma and correlated with advanced disease and poor prognosis [[19], [20], [21], [22]]. HER4 expression has not been examined as extensively and its prognostic value is not known.

Disruption of intracellular communications between stromal and cancer cells could affect tumor proliferation and represents an interesting therapeutic strategy. In this context, we hypothesized that targeting NRG, which we here show to be secreted both by CAFs and tumor cells, could be a novel therapy to overcome CAF-triggered tumor growth in pancreatic cancer. Here, we investigated, in in vivo and in vitro co-culture of pancreatic cancer cells and CAFs, the therapeutic potential of an original monoclonal antibody (7E3) that targets NRG1 Ig-like domain. We show promising tumor growth inhibitory effects of this antibody which promotes antibody dependent cellular cytotoxicity (ADCC) in NRG1-permeated pancreatic cancer cells and in NRG1-positive CAFs and blocks NRG1-mediated HER3 activation and downstream signaling pathway induction.

Section snippets

Cell lines and reagents

The BxPC-3, CFPAC-1, HPAC, AsPC-1, SW1990, MiaPaCa-2 and MCF7 cancer cell lines were obtained from ATCC (Rockville, MD, USA) and cultured following the ATCC recommendations. The identity of cell lines was confirmed by genetics authentification test (Eurofins, Ebersberg). BxPC-3-Luc, HPAC-Luc, shHER3 BxPC-3 and shluc BxPC-3 cells were generated previously in the laboratory [23]. Stably-transfected AsPC-1-Mock (vector alone) and AsPC-1-NRG1 cells were obtained as previously described for

NRG1 regulates pancreatic tumor growth in an autocrine and paracrine manner

RT-qPCR expression analysis showed NRG1 expression in 17/33 (51.5%) pancreatic tumor samples (Fig. 1A). Then, NRG1β1 gene expression was assessed in five KRAS-mutated pancreatic cancer cell lines and one KRAS-wild type cell line (BxPC-3). BxPC3, CFPAC and SW1990 cells expressed NRG1, whereas HPAC, MiaPaca2 and AsPC-1 did not (Fig. 1B). As positive control, we developed a NRG1 positive cell derived of AsPC-1 (AsPC-1-NRG1 cells). As negative control, we used normal pancreatic cells (NPC). By

Discussion

The 7E3 antibody blocked NRG1-dependent tumor cell growth by decreasing HER3 activity and expression level and promoting ADCC activation against tumors cells and micro-environment. This mechanism can contribute to the therapeutic effect observed in mice and can be explained by its original binding. Indeed, our antibody exhibited a high degree of specificity and affinity for NRG1 IgG-like domain, and did not prevent the NRG1 binding to HER3. This explains 7E3 ability to target NRG1 already

Funding

This work was supported by grants from the Ligue Nationale Contre le Cancer-Comité de l’Hérault, France, from the LabEx MabImprove (ANR-10-LABX-53-01), France, from the SIRIC Montpellier Cancer, “INCa-DGOS-INSERM 6045”, France, from the Cancéropole Grand Sud Ouest, France.

Conflicts of interest

Charline Ogier, Christel Larbouret, Thierry Chardès and André Pèlegrin are inventors in the anti-NRG1 mAb patents related to this work. The other authors declare no conflict of interest.

Acknowledgments

We thank S. Bousquié, for technical assistance and N. Robin for providing pancreatic tumors. We thank the Montpellier RIO Imaging and the RHEM histology facilities.

References (50)

  • M.F. Bijlsma et al.

    The conflicting roles of tumor stroma in pancreatic cancer and their contribution to the failure of clinical trials: a systematic review and critical appraisal

    Canc. Metastasis Rev.

    (2015)
  • J.J. Lee et al.

    Stromal response to Hedgehog signaling restrains pancreatic cancer progression

    Proc. Natl. Acad. Sci. U. S. A

    (2014)
  • R. Kalluri

    The biology and function of fibroblasts in cancer

    Nat. Rev. Canc.

    (2016)
  • D. Öhlund et al.

    Fibroblast heterogeneity in the cancer wound

    J. Exp. Med.

    (2014)
  • J.C. Montero et al.

    Neuregulins and cancer

    Clin. Canc. Res.

    (2008)
  • G. Qian et al.

    Heregulin and HER3 are prognostic biomarkers in oropharyngeal squamous cell carcinoma

    Cancer

    (2015)
  • A. Kolb et al.

    Expression and differential signaling of heregulins in pancreatic cancer cells

    Int. J. Canc.

    (2007)
  • M.S. Tsai et al.

    Blockage of heregulin expression inhibits tumorigenicity and metastasis of breast cancer

    Oncogene

    (2003)
  • H. Lee et al.

    A naturally occurring secreted human ErbB3 receptor isoform inhibits heregulin-stimulated activation of ErbB2, ErbB3, and ErbB4

    Canc. Res.

    (2001)
  • H. Kawakami et al.

    The anti-HER3 antibody patritumab abrogates cetuximab resistance mediated by heregulin in colorectal cancer cells

    Oncotarget

    (2014)
  • H. Friess et al.

    Enhanced erbB-3 expression in human pancreatic cancer correlates with tumor progression

    Clin. Canc. Res.

    (1995)
  • J.S. Liles et al.

    ErbB3 expression promotes tumorigenesis in pancreatic adenocarcinoma

    Canc. Biol. Ther.

    (2010)
  • Y. Yamanaka et al.

    Coexpression of epidermal growth factor receptor and ligands in human pancreatic cancer is associated with enhanced tumor aggressiveness

    Anticancer Res.

    (1993)
  • H. Safran et al.

    Overexpression of the HER-2/neu oncogene in pancreatic adenocarcinoma

    Am. J. Clin. Oncol.

    (2001)
  • G. Thomas et al.

    HER3 as biomarker and therapeutic target in pancreatic cancer: new insights in pertuzumab therapy in preclinical models

    Oncotarget

    (2014)
  • Cited by (39)

    • Targeting HER3-dependent activation of nuclear AKT improves radiotherapy of non-small cell lung cancer

      2022, Radiotherapy and Oncology
      Citation Excerpt :

      Likewise, PI3K/AKT signaling in association with enhanced HER3 expression was shown to be the underlying mechanism of resistance to EGFR TK inhibitors in NSCLC [46]. Moreover, a study by Ogier et al. demonstrated that targeting of NRG-HER3 axis using NRG neutralizing antibody is an effective therapeutic approach for pancreatic carcinoma [47]. In the present study we showed that anti-EGFR antibody cetuximab does not block AKT activity after irradiation in lung cancer cells.

    • Cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs); where do they stand in tumorigenesis and how they can change the face of cancer therapy?

      2022, European Journal of Pharmacology
      Citation Excerpt :

      Scriptaid, a selective inhibitor of HDACs 1/3/8, also reported to could suppress TGFβ-mediated CAF differentiation, reduce ECM secretion, and impair cell invasion in murine model of melanoma (Kim et al., 2018). Targeting neuregulin (NRG1), a highly expressed molecules on CAFs, using antibodies such as 7E3 is another mechanism that could reduce the activity of CAFs in pancreatic tumors (Ogier et al., 2018). MicroRNAs are another tool that could be recruited to shut down the activity of CAFs in TME.

    View all citing articles on Scopus
    View full text