Elsevier

Clinical Lung Cancer

Volume 19, Issue 3, May 2018, Pages e277-e285
Clinical Lung Cancer

Original Study
Continuous Infusion of Cilengitide Plus Chemoradiotherapy for Patients With Stage III Non–Small-cell Lung Cancer: A Phase I Study

https://doi.org/10.1016/j.cllc.2017.11.002Get rights and content

Abstract

Introduction

Because of our previous preclinical results, we conducted a phase I study associating the specific αvβ3/αvβ5 integrin inhibitor cilengitide, given as a continuous infusion, with exclusive chemoradiotherapy for patients with stage III non–small-cell lung cancer.

Patients and Methods

A standard 3+3 dose escalation design was used. Cilengitide was given as a continuous infusion (dose levels of 12, 18, 27, and 40 mg/h), starting 2 weeks before and continuing for the whole course of chemoradiotherapy (66 Gy combined with platinum/vinorelbine), and then at a dose of 2000 mg twice weekly in association with chemotherapy. 2-Deoxy-2-[fluorine-18]fluoro-d-glucose positron emission tomography (PET) and computed tomography scans were performed before and after the first 2 weeks of cilengitide administration and then every 3 months.

Results

Of the 14 patients included, 11 were evaluable for evaluation of the dose-limiting toxicities (DLTs). One DLT, a tracheobronchial fistula, was reported with the 40 mg/h dose. No relevant adverse events related to cilengitide were observed overall. At the PET evaluation 2 months after chemoradiotherapy, 4 of 9 patients had a complete response and 4 had a partial response. The median progression-free and overall survival was 14.4 months (95% confidence interval [CI], 8.4 to not reached) and 29.4 months (95% CI, 11.73 to not reached), respectively.

Conclusion

Cilengitide, given continuously with chemoradiotherapy, showed acceptable toxicity and gave encouraging clinical results.

Introduction

Despite recent advances in the planning and delivery of radiation therapy for unresectable stage III non–small-cell lung cancer (NSCLC), the therapeutic index has not been further improved owing to physical limitations and dose-limiting structures, as recently shown by unsuccessful dose-escalating studies.1 For additional progress to occur, technological innovations must be complemented by biologic innovations, such as the development of novel radiosensitizing agents and biology-driven patient selection.

Based on preclinical data, which demonstrated the role of fibroblast growth factor (FGF-2) and αvβ3 integrin in radioresistance and hypoxia,2, 3, 4, 5 we investigated the predictive value of FGF-2 and αvβ3 integrin expression in NSCLC patients treated with chemoradiotherapy. The results showed that the combined expression of FGF-2 and β3 integrin was related to local relapse, thus confirming the role of these factors in tumor radiosensitivity.6

The integrin receptors, αvβ3 and αvβ5, whose presence on both endothelial and tumor cells are crucial for continued cell survival, represent attractive targets for antiangiogenesis strategies and radiosensitization.7, 8, 9 Tumor radiosensitivity is modulated by both tumor cell and endothelial cell pathways.10

Cilengitide (EMD121974; Merck KGaA, Darmstadt, Germany), the inner salt of a cyclized RGD pentapeptide (cyclo-[Arg-Gly-Asp-Dphe-(NMeVal)]), which has been shown to radiosensitize lung cancer cells and endothelial cells in vitro,11 is a potent and selective αvβ3 and αvβ5 antagonist.12 Inhibiting αvβ3 and αvβ5 integrins in turn counteracts the endothelial cell invasion and differentiation induced by vascular endothelial growth factor (VEGF)-A or FGF, leading to an antiangiogenic effect.13 Moreover, it has been shown in a glioma model that inhibition of αvβ3 or αvβ5 integrins in hypoxic cells decreases the hypoxia-inducible factor 1α intracellular level, which also explains the antiangiogenic effects of cilengitide.5

Cilengitide has inhibited tumor growth in a dose-dependent manner in numerous in vivo systems.14, 15, 16 Regarding its combination with chemotherapy, cilengitide exerted in vitro and in vivo synergistic antiproliferative action in preclinical studies with different cytotoxic agents,17, 18, 19 with promising results for the association in early clinical trials, especially with cisplatin.20, 21 Several phase I and II studies, conducted on adult patients with advanced solid tumors, have used cilengitide, given as a 1-hour infusion twice weekly, with a good safety profile.22, 23, 24 A randomized phase II study also tested cilengitide at 2000 mg twice weekly or 2000 mg for 5 d/wk combined with temozolomide and radiotherapy to treat glioblastoma.25 Only one of the reported dosing strategies for cilengitide in humans has used continuous administration.26 Most have used intermittent schedules, including within the disappointing phase III CENTRIC study (cilengitide, temozolomide, and radiation therapy in treating patients with newly diagnosed glioblastoma and methylated gene promoter status) of glioblastoma.27 However, cilengitide has a short plasma half-life of 3 to 5 hours, supporting the idea that continuous exposure to cilengitide might be necessary for its optimal efficacy and, thus, warrants investigation.

We designed and conducted a phase I clinical study that combined cilengitide, given as a continuous infusion, with exclusive and concomitant chemoradiotherapy for stage III NSCLC patients.

Section snippets

Eligibility Criteria

Eligible patients had to satisfy the following criteria: age > 18 years, inoperable stage IIIA or IIIB NSCLC (American Joint Committee on Cancer, 7th edition),28 World Health Organization performance status of 0 to 1, and combined modality therapy deemed appropriate by a multidisciplinary tumor board. Initial staging included chest computed tomography (CT), 2-deoxy-2-[fluorine-18]fluoro-d-glucose (18F-FDG) positron emission tomography (PET), and brain magnetic resonance imaging. Other

Patients and Treatment Administrations

From March 2010 to July 2013, 14 patients were included in the present study. These included 5 patients at level 0 (12 mg/h), 3 at level 1 (18 mg/h), 3 at level 2 (27 mg/h), and 3 at level 3 (40 mg/h). Of the 14 patients, 3 were considered not evaluable for DLT and efficacy owing to early withdrawal (patients 1 and 14 were withdrawn because of metastatic disease found on imaging studies performed after 2 weeks of exclusive cilengitide treatment; patient 2 was withdrawn because of cholestatic

Discussion

In the present open-label dose-escalation study that evaluated cilengitide administered as a continuous infusion during concurrent chemoradiotherapy for stage III NSCLC, no DLT was observed among 11 evaluable patients until a level of 40 mg/h was reached. One DLT, a tracheobronchial fistula, was reported at the third-level dose (ie, cilengitide at 40 mg/h). As reported in previous studies of cilengitide, when given intermittently22, 23, 25, 27 or continuously,26 overall, this drug has been well

Disclosure

E.C.J.M. served as an advisory board member on Cilengitide. The remaining authors declare that they have no competing interests.

Acknowledgments

This work received a grant from Merck-Serono. Merck Serono had no role in the trial design, collection and interpretation of data, or in the writing of this article. Merck Serono performed a scientific review of our report; however, the views and opinions do not necessarily reflect those of Merck Serono.

References (51)

  • K. Dittmann et al.

    Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNA-dependent protein kinase

    J Biol Chem

    (2005)
  • N. Ready et al.

    Chemoradiotherapy and gefitinib in stage III non-small cell lung cancer with epidermal growth factor receptor and KRAS mutation analysis: Cancer and Leukemia Group B (CALEB) 30106, a CALGB-stratified phase II trial

    J Thorac Oncol

    (2010)
  • S. Rothschild et al.

    Gefitinib in combination with irradiation with or without cisplatin in patients with inoperable stage III non-small cell lung cancer: a phase I trial

    Int J Radiat Oncol Biol Phys

    (2011)
  • R. Komaki et al.

    Adding erlotinib to chemoradiation improves overall survival but not progression-free survival in stage III non-small cell lung cancer

    Int J Radiat Oncol Biol Phys

    (2015)
  • M.S. O'Reilly et al.

    Endostatin: an endogenous inhibitor of angiogenesis and tumor growth

    Cell

    (1997)
  • A. Abdollahi et al.

    Endostatin's antiangiogenic signaling network

    Mol Cell

    (2004)
  • E. Cohen-Jonathan et al.

    Radioresistance induced by the high molecular forms of the basic fibroblast growth factor is associated with an increased G2 delay and a hyperphosphorylation of p34CDC2 in HeLa cells

    Cancer Res

    (1997)
  • I. Ader et al.

    RhoB controls the 24 kDa FGF-2-induced radioresistance in HeLa cells by preventing post-mitotic cell death

    Oncogene

    (2002)
  • S. Monferran et al.

    Alphavbeta3 and alphavbeta5 integrins control glioma cell response to ionising radiation through ILK and RhoB

    Int J Cancer

    (2008)
  • N. Skuli et al.

    Alphavbeta3/alphavbeta5 integrins-FAK-RhoB: a novel pathway for hypoxia regulation in glioblastoma

    Cancer Res

    (2009)
  • M. Friedlander et al.

    Definition of two angiogenic pathways by distinct alpha v integrins

    Science

    (1995)
  • M. Trikha et al.

    Multiple roles for platelet GPIIb/IIIa and alphavbeta3 integrins in tumor growth, angiogenesis, and metastasis

    Cancer Res

    (2002)
  • M. Garcia-Barros et al.

    Tumor response to radiotherapy regulated by endothelial cell apoptosis

    Science

    (2003)
  • J.-P. Xiong et al.

    Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand

    Science

    (2002)
  • R.E. Nisato et al.

    Alphav beta 3 and alphav beta 5 integrin antagonists inhibit angiogenesis in vitro

    Angiogenesis

    (2003)
  • Cited by (18)

    • Metastasis prevention: How to catch metastatic seeds

      2023, Biochimica et Biophysica Acta - Reviews on Cancer
    View all citing articles on Scopus

    C.M. and J.K. contributed equally to this report and are co-first authors.

    J.M. and E.C.-J.M. are co-last authors.

    View full text