A Phase II, Randomized, Double-Blind, Placebo-Controlled Study of Simtuzumab in Combination with FOLFIRI for the Second-Line Treatment of Metastatic KRAS Mutant Colorectal Adenocarcinoma

J Randolph Hecht, Al B Benson 3rd, Dmitry Vyushkov, Yingsi Yang, Johanna Bendell, Udit Verma, J Randolph Hecht, Al B Benson 3rd, Dmitry Vyushkov, Yingsi Yang, Johanna Bendell, Udit Verma

Abstract

Lessons learned: The safety profile in the patient groups who received FOLFIRI and simtuzumab did not differ from that in the FOLFIRI and placebo group.The addition of simtuzumab to chemotherapy with FOLFIRI does not improve clinical outcomes in patients with metastatic KRAS mutant colorectal carcinoma.

Background: Simtuzumab, a humanized IgG4 monoclonal antibody to lysyl oxidase-like 2 (LOXL2), blocks desmoplastic reaction in colorectal carcinoma (CRC) cells in vitro.

Methods: Patients with metastatic Kirsten rat sarcoma viral oncogene homolog (KRAS) mutant CRC were randomized to receive second-line 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI) with either 200 or 700 mg simtuzumab or placebo every 2 weeks in cycles of 28 days. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and safety were assessed.

Results: In total, 249 patients were randomized and treated with FOLFIRI/simtuzumab 700 mg (n = 84), FOLFIRI/simtuzumab 200 mg (n = 85), and FOLFIRI/placebo (n = 80). After a median follow-up of 5.1, 3.8, and 5.5 months, respectively, median PFS for each of the respective treatment groups was 5.5 months (adjusted HR [95% CI], p value versus placebo; 1.32 [0.92, 1.89]; p = .10), 5.4 months (1.45 [1.01, 2.06]; p = .04), and 5.8 months. Median OS was 11.4 months (1.23 [0.80, 1.91]; p = .25), 10.5 months (1.50 [0.98, 2.30]; p = .06), and 16.3 months, respectively. ORR was 11.9%, 5.9%, and 10%, respectively. Simtuzumab was tolerable in metastatic KRAS mutant CRC patients.

Conclusion: The addition of simtuzumab to FOLFIRI did not improve clinical outcomes in patients with metastatic KRAS mutant CRC. The Oncologist 2017;22:243-e8.

Trial registration: ClinicalTrials.gov NCT01479465.

© AlphaMed Press; the data published online to support this summary is the property of the authors.

Figures

Figure 1.
Figure 1.
Stratified Kaplan–Meier plot of progression‐free survival by investigator assessment (FAS population). Abbreviations: CI, confidence interval; FAS, full analysis set (all patients randomized and treated with ≥1 dose of study drug); HR, hazard ratio (numbers in brackets are 95% CIs); PFS, progression‐free survival; SIM, simtuzumab.
Figure 2.
Figure 2.
Stratified Kaplan–Meier plot of overall survival (FAS population). Abbreviations: CI, confidence interval; FAS, full analysis set (all patients randomized and treated with ≥1 dose of study drug); HR, hazard ratio (numbers in brackets are 95% CIs); OS, overall survival; SIM, simtuzumab.

References

    1. Ferlay J, Soerjomataram I, Dikshit R et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359–E386.
    1. Armaghany T, Wilson JD, Chu Q et al. Genetic alterations in colorectal cancer. Gastrointest Cancer Res 2012;5:19–27.
    1. Benvenuti S, Sartore‐Bianchi A, Di Nicolantonio F et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti‐epidermal growth factor receptor antibody therapies. Cancer Res 2007;67:2643–2648.
    1. Phipps AI, Buchanan DD, Makar KW et al. KRAS‐mutation status in relation to colorectal cancer survival: The joint impact of correlated tumour markers. Br J Cancer 2013;108:1757–1764.
    1. NCCN Clinical Practice Guidelines in Oncology. Pancreatic Adenocarcinoma. 2016. Available at . Accessed May 12, 2016.
    1. Bremnes RM, Dønnem T, Al‐Saad S et al. The role of tumor stroma in cancer progression and prognosis: Emphasis on carcinoma‐associated fibroblasts and non‐small cell lung cancer. J Thorac Oncol 2011;6:209–217.
    1. Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature 2004;432:332–337.
    1. Fong SF, Dietzsch E, Fong KS et al. Lysyl oxidase‐like 2 expression is increased in colon and esophageal tumors and associated with less differentiated colon tumors. Genes Chromosomes Cancer 2007;46:644–655.
    1. Barry‐Hamilton V, Spangler R, Marshall D et al. Allosteric inhibition of lysyl oxidase‐like‐2 impedes the development of a pathologic microenvironment. Nat Med 2010;16:1009–1017.
    1. Zaffryar‐Eilot S, Marshall D, Voloshin T et al. Lysyl oxidase‐like‐2 promotes tumour angiogenesis and is a potential therapeutic target in angiogenic tumours. Carcinogenesis 2013;34:2370–2379.
    1. Payne SL, Hendrix MJ, Kirschmann DA. Paradoxical roles for lysyl oxidases in cancer–a prospect. J Cell Biochem 2007;101:1338–1354.
    1. Hecht JR, Bendell JC, Vyushkov D et al. A phase II, randomized, double‐blinded, placebo‐controlled study of simtuzumab or placebo in combination with FOLFIRI for the second line treatment of metastatic KRAS mutant colorectal adenocarcinoma. J Clin Oncol 2015;33:abstr 3537.
    1. Rodriguez HM, Vaysberg M, Mikels A et al. Modulation of lysyl oxidase‐like 2 enzymatic activity by an allosteric antibody inhibitor. J Biol Chem 2010;285:20964–20974.
    1. Van Bergen T, Marshall D, Van de Veire S et al. The role of LOX and LOXL2 in scar formation after glaucoma surgery. Invest Ophthalmol Vis Sci 2013;54:5788–5796.
    1. Peng L, Ran YL, Hu H et al. Secreted LOXL2 is a novel therapeutic target that promotes gastric cancer metastasis via the Src/FAK pathway. Carcinogenesis 2009;30:1660–1669.
    1. LoRusso P, Hecht J, Thai D et al. Phase I and IIa studies of simtuzumab alone and in combination with FOLFIRI in patients with advanced solid tumors. J Clin Oncol 2014;32:abstr 554.

Source: PubMed

3
Prenumerera