Review
Resistance to sunitinib in renal cell carcinoma: From molecular mechanisms to predictive markers and future perspectives

https://doi.org/10.1016/j.bbcan.2014.11.002Get rights and content

Highlights

  • Sunitinib resistance in renal cell carcinoma appears to be transient and reversible.

  • Tumor hypoxia plays a crucial role in several resistance mechanisms.

  • Hypoxia-mediated cMET signaling leads to an increased malignant potential.

  • Targeting cMET seems to be the most promising pharmacological treatment option.

  • DNA promoter hypermethylation could represent promising predictive factors.

Abstract

The introduction of agents that inhibit tumor angiogenesis by targeting vascular endothelial growth factor (VEGF) signaling has made a significant impact on the survival of patients with metastasized renal cell carcinoma (RCC). Sunitinib, a tyrosine kinase inhibitor of the VEGF receptor, has become the mainstay of treatment for these patients. Although treatment with sunitinib substantially improved patient outcome, the initial success is overshadowed by the occurrence of resistance. The mechanisms of resistance are poorly understood. Insight into the molecular mechanisms of resistance will help to better understand the biology of RCC and can ultimately aid the development of more effective therapies for patients with this infaust disease. In this review we comprehensively discuss molecular mechanisms of resistance to sunitinib and the involved biological processes, summarize potential biomarkers that predict response and resistance to treatment with sunitinib, and elaborate on future perspectives in the treatment of metastasized RCC.

Introduction

Renal cell carcinoma (RCC), which arises from the renal parenchyma, is the most common kidney cancer subtype, accounting for approximately 90% of all cases [1], [2]. Of the patients diagnosed with RCC, 20–30% present with metastasized disease, and another ~ 30% of the patients treated for localized disease develop metastases during follow-up [3]. RCC is not a single entity, but comprises a heterogeneous group of malignancies, of which clear cell RCC (ccRCC) is the most common (75–80%) and the best studied to date. ccRCCs are highly vascularized tumors that are characterized by frequent inactivation (50–75%) of the Von Hippel–Lindau (VHL) gene [2], [4], [5]. The product of the VHL gene, pVHL, plays an important role in down-regulating the expression of the hypoxia inducible factor 1 (HIF1) transcription factor, which leads to decreased angiogenesis (Fig. 1). Inactivation of pVHL, e.g. by mutation, deletion or promoter CpG island methylation of the VHL gene, leads to accumulation of HIF1 and increased transcription of HIF1 target genes e.g. vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). The frequent inactivation of VHL provided the rationale for the development of antiangiogenic drugs to treat ccRCC such as sunitinib, pazopanib, sorafenib, and axitinib [4]. Sunitinib is an oral multiple tyrosine kinases inhibitor (TKI), that inhibits the family of vascular endothelial growth factor receptors (VEGFR-1, VEGFR-2 and VEGFR-3), the platelet-derived growth factor receptors (PDGFRα and PDGFRβ), FLT3, cKIT and RET. The inhibitory effect of sunitinib on tumor angiogenesis is mainly achieved by blocking VEGFR and PDGFR, two key players in the pathogenesis of ccRCC [6], [7]. In metastasized ccRCC, sunitinib treatment resulted in a significant longer progression free survival (PFS) (11 versus 5 months, P < 0.001) and overall survival (OS) (26.4 versus 21.8 months, P = 0.049) compared to treatment with interferon alpha (IFN-α) [8], and it has therefore become the mainstay of treatment.

Initial response rates to sunitinib lie between 30% and 40% [9], [10], [11]. However, disease progression usually occurs after a median of 6–15 months [12], indicating the existence of intrinsic (primary) and/or acquired (secondary) resistance. The resistance mechanisms to sunitinib can be divided in several distinct groups, namely up-regulation of proangiogenic signaling pathways, resistance mediated by the tumor microenvironment, increased tumor invasiveness and metastasis, activation of alternative signaling pathways, inadequate target inhibition, and resistance mediated by the action of microRNAs (Fig. 2). More recently, it was shown that pazopanib has similar efficacy as sunitinib, but with a favorable safety and toxicity profile [13], [14]. As a result, sunitinib and pazopanib are used interchangeably in clinical practice. It is likely that the potential resistance mechanisms that are identified for sunitinib may also account for pazopanib, since they have the same molecular targets. However, to the best of our knowledge, no literature on resistance to pazopanib has yet been published. Therefore, the focus of this review will be on sunitinib resistance. We will discuss data from preclinical and clinical studies on mechanisms of sunitinib resistance, and summarize the current knowledge on potential predictive markers that can help to select patients that are likely to benefit from treatment with sunitinib. Finally, we will discuss future perspectives on how to optimize the treatment of patients with metastasized RCC.

Section snippets

Up-regulation of proangiogenic pathways

An established cause for the development of resistance to VEGF-targeted therapy is tumor hypoxia. Tumors exceeding a volume of 1 mm3 usually contain regions of hypoxia [15]. It is well known that tumor hypoxia is associated with increased invasiveness and metastasis, and poorer patient survival [15], [16]. Targeting tumor angiogenesis, e.g. by blocking VEGF signaling, leads to increased hypoxia in the tumor which in turn leads to necrosis and decrease of tumor burden. However, hypoxia also leads

Potential predictive biomarkers of response and resistance to sunitinib

As described above, a subgroup of RCC patients is intrinsically resistant to, or develops acquired resistance upon, treatment with sunitinib. (Bio)markers accurately identifying these patients will help managing this disease more optimally.

Conclusions and future perspectives

Although different mechanisms of resistance have been identified such as up-regulation of proangiogenic pathways, recruitment of BMDCs, promotion of tumor invasiveness and metastasis, and activation of alternative signaling pathways, one factor seems to play a crucial role in all these processes: tumor hypoxia. Targeting the tumor vasculature with VEGF-targeted agents renders the tumor cells hypoxic with subsequent accumulation of HIF1, which in turn leads to up-regulation of proangiogenic

References (176)

  • R.J. Motzer et al.

    Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial

    Lancet Oncol.

    (2014)
  • M. Schmidinger

    Third-line dovitinib in metastatic renal cell carcinoma

    Lancet Oncol.

    (2014)
  • D. Martin et al.

    CXCL8/IL8 stimulates vascular endothelial growth factor (VEGF) expression and the autocrine activation of VEGFR2 in endothelial cells by activating NFkB trough the CBM (Carma3/Bcl10/Malt1) complex

    J. Biol. Chem.

    (2009)
  • D. Ribatti

    The controversial role of placental growth factor in tumor growth

    Cancer Lett.

    (2011)
  • C. Parr et al.

    Placenta growth factor is over-expressed and has prognostic value in human breast cancer

    Eur. J. Cancer

    (2005)
  • C.N. Chen et al.

    The significance of placenta growth factor in angiogenesis and clinical outcome of human gastric cancer

    Cancer Lett.

    (2004)
  • C. Fischer et al.

    Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels

    Cell

    (2007)
  • A. Eriksson et al.

    Placenta growth factor-1 antagonizes VEGF-induced angiogenesis and tumor growth by the formation of functionally inactive PlGF-1/VEGF heterodimers

    Cancer Cell

    (2002)
  • Y.J. Koh et al.

    Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage

    Cancer Cell

    (2010)
  • C. Mailhos et al.

    Delta4, an endothelial specific notch ligand expressed at sites of physiological and tumor angiogenesis

    Differentiation

    (2001)
  • F. Shojaei et al.

    Role of the microenvironment in tumor growth and in refractoriness/resistance to anti-angiogenic therapies

    Drug Resist. Updat.

    (2008)
  • Y. Crawford et al.

    Tumor and stromal pathways medating refractoriness/resistance to anti-angiogenic therapies

    Trends Pharmacol. Sci.

    (2009)
  • R. Du et al.

    HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion

    Cancer Cell

    (2008)
  • J. Condeelis et al.

    Macrophages: obligate partners for tumor cell migration, invasion, and metastasis

    Cell

    (2006)
  • L. Yang et al.

    Expansion of myeloid immune suppressor Gr + CD11b + cells in tumor-bearing host directly promotes tumor angiogenesis

    Cancer Cell

    (2004)
  • Y. Crawford et al.

    PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment

    Cancer Cell

    (2009)
  • J.M.L. Ebos et al.

    Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis

    Cancer Cell

    (2009)
  • M. Paez-Ribes et al.

    Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis

    Cancer Cell

    (2009)
  • J.R. Gnarra et al.

    Mutations of the VHL tumour suppressor gene in renal carcinoma

    Nat. Genet.

    (1994)
  • R.E. Banks et al.

    Genetic and epigenetic analysis of von Hippel–Lindau (VHL) gene alterations and relationship with clinical variables in sporadic renal cancer

    Cancer Res.

    (2006)
  • S. Faivre et al.

    Molecular basis for sunitinib efficacy and future clinical development

    Nat. Rev. Drug Discov.

    (2007)
  • D. Huang et al.

    Sunitinib acts primarily on tumor endothelium rather than tumor cells to inhibit the growth of renal cell carcinoma

    Cancer Res.

    (2010)
  • R.J. Motzer et al.

    Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma

    J. Clin. Oncol.

    (2009)
  • R.J. Motzer et al.

    Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma

    J. Clin. Oncol.

    (2006)
  • R.J. Motzer et al.

    Sunitinib in patients with metastatic renal cell carcinoma

    JAMA

    (2006)
  • R.J. Motzer et al.

    Pazopanib versus sunitinib in metastatic renal-cell carcinoma

    N. Engl. J. Med.

    (2013)
  • B. Escudier et al.

    Randomized, controlled, double-blind, cross-over trial assessing treatment preference for pazopanib versus sunitinib in patients with metastatic renal cell carcinoma: PISCES Study

    J. Clin. Oncol.

    (2014)
  • A.L. Harris

    Hypoxia—a key regulatory factor in tumour growth

    Nat. Rev. Cancer

    (2002)
  • M.M. Baldewijns et al.

    VHL and HIF signaling in renal cell carcinogenesis

    J. Pathol.

    (2010)
  • J.D. Sun et al.

    Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibitis tumor growth in preclinical models of cancer

    Clin. Cancer Res.

    (2012)
  • F. Meng et al.

    Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302

    Mol. Cancer Ther.

    (2012)
  • S. Portwood et al.

    Activity of the hypoxia-activated prodrug, TH-302, in preclinical human acute myeloid leukemia (AML) models

    Clin. Cancer Res.

    (2013)
  • J. Hu et al.

    Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro

    Mol. Cancer Ther.

    (2013)
  • Q. Liu et al.

    TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules

    Cancer Chemother. Pharmacol.

    (2012)
  • K.N. Ganjoo et al.

    A phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma

    Oncology

    (2011)
  • G.J. Weiss et al.

    Phase 1 study of the safety, tolerability, and pharmacokinetics of TH-302, a hypoxia-activated prodrug, in patients with advanced solid malignancies

    Clin. Cancer Res.

    (2011)
  • J.C. Welti et al.

    Fibroblast growth factor 2 regulates endothelial cell sensitivity to sunitinib

    Oncogene

    (2011)
  • B. Escudier et al.

    Sorafenib in advanced clear-cell renal-cell carcinoma

    N. Engl. J. Med.

    (2007)
  • D. Huang et al.

    Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma

    Cancer Res.

    (2010)
  • Y. Mizukami et al.

    Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells

    Nat. Med.

    (2005)
  • Cited by (93)

    • New frontiers against sorafenib resistance in renal cell carcinoma: From molecular mechanisms to predictive biomarkers

      2021, Pharmacological Research
      Citation Excerpt :

      NS-398, an inhibitor of HIF-2α/COX-2 pathway, can reverse hypoxia induced sorafenib resistance in RCC [58]. Furthermore, a series of preclinical and clinical data have also confirmed that HAPs, which cause DNA damage only under extreme hypoxic conditions, have a good application prospect in the treatment of RCC patients who are resistant to sorafenib therapy [16,64]. Meanwhile, nutlin-3, an inhibitor of MDM2-p53 interaction, can synergistically enhance the antitumor efficacy of sorafenib by inhibiting the ubiquitin degradation of p53 and activating the p53-regulated apoptotic pathway [65].

    View all citing articles on Scopus
    View full text