Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer

Daniel V T Catenacci, Melissa R Junttila, Theodore Karrison, Nathan Bahary, Margit N Horiba, Sreenivasa R Nattam, Robert Marsh, James Wallace, Mark Kozloff, Lakshmi Rajdev, Deirdre Cohen, James Wade, Bethany Sleckman, Heinz-Josef Lenz, Patrick Stiff, Pankaj Kumar, Peng Xu, Les Henderson, Naoko Takebe, Ravi Salgia, Xi Wang, Walter M Stadler, Frederic J de Sauvage, Hedy L Kindler, Daniel V T Catenacci, Melissa R Junttila, Theodore Karrison, Nathan Bahary, Margit N Horiba, Sreenivasa R Nattam, Robert Marsh, James Wallace, Mark Kozloff, Lakshmi Rajdev, Deirdre Cohen, James Wade, Bethany Sleckman, Heinz-Josef Lenz, Patrick Stiff, Pankaj Kumar, Peng Xu, Les Henderson, Naoko Takebe, Ravi Salgia, Xi Wang, Walter M Stadler, Frederic J de Sauvage, Hedy L Kindler

Abstract

Purpose: Sonic hedgehog (SHH), an activating ligand of smoothened (SMO), is overexpressed in > 70% of pancreatic cancers (PCs). We investigated the impact of vismodegib, an SHH antagonist, plus gemcitabine (GV) or gemcitabine plus placebo (GP) in a multicenter phase Ib/randomized phase II trial and preclinical PC models.

Patients and methods: Patients with PC not amenable to curative therapy who had received no prior therapy for metastatic disease and had Karnofsky performance score ≥ 80 were enrolled. Patients were randomly assigned in a one-to-one ratio to GV or GP. The primary end point was progression-free-survival (PFS). Exploratory correlative studies included serial SHH serum levels and contrast perfusion computed tomography imaging. To further investigate putative biologic mechanisms of SMO inhibition, two autochthonous pancreatic cancer models (Kras(G12D); p16/p19(fl/fl); Pdx1-Cre and Kras(G12D); p53(R270H/wt); Pdx1-Cre) were studied.

Results: No safety issues were identified in the phase Ib portion (n = 7), and the phase II study enrolled 106 evaluable patients (n = 53 in each arm). Median PFS was 4.0 and 2.5 months for GV and GP arms, respectively (95% CI, 2.5 to 5.3 and 1.9 to 3.8, respectively; adjusted hazard ratio, 0.81; 95% CI, 0.54 to 1.21; P = .30). Median overall survival (OS) was 6.9 and 6.1 months for GV and GP arms, respectively (95% CI, 5.8 to 8.0 and 5.0 to 8.0, respectively; adjusted hazard ratio, 1.04; 95% CI, 0.69 to 1.58; P = .84). Response rates were not significantly different. There were no significant associations between correlative markers and overall response rate, PFS, or OS. Preclinical trials revealed no significant differences with vismodegib in drug delivery, tumor growth rate, or OS in either model.

Conclusion: The addition of vismodegib to gemcitabine in an unselected cohort did not improve overall response rate, PFS, or OS in patients with metastatic PC. Our preclinical and clinical results revealed no statistically significant differences with respect to drug delivery or treatment efficacy using vismodegib.

Trial registration: ClinicalTrials.gov NCT01064622.

Conflict of interest statement

Authors' disclosures of potential conflicts of interest are found in the article online at www.jco.org. Author contributions are found at the end of this article.

© 2015 by American Society of Clinical Oncology.

Figures

Fig 1.
Fig 1.
CONSORT diagram of clinical trial enrollment and treatment in phase II trial. gem, gemcitabine.
Fig 2.
Fig 2.
(A) Progression-free and (B) overall survival by treatment arm. Blue, gemcitabine plus vismodegib; gold, gemcitabine plus placebo. Hazard ratio (HR) after adjusting (adj) for Karnofsky performance score and disease status (newly diagnosed v recurrent).
Fig 3.
Fig 3.
Clinical trial translational correlatives. Serum SHH levels (A) comparing controls (n = 40) with patients with pancreatic cancer enrolled onto trial (n = 89), (B) by treatment group (gemcitabine plus vismodegib [GV] or gemcitabine plus placebo [GP]) with increasing treatment (TX) cycle, and association with age in (C) patients with cancer and (D) controls. (E) Radiologic correlatives evaluating association of baseline tumor perfusion with tumor response to therapy.
Fig 4.
Fig 4.
Effects of hedgehog pathway antagonism (HhAntag) on vasculature, stromal content, and intratumoral gemcitabine metabolites in KrasG12D; p16/p19fl/fl; Pdx1-Cre (KPP) tumors. (A) Quantitation of immunohistochemical staining for meca-32 expression in pancreatic tumors of KPP mice treated with vehicle (circles; n = 13) or smoothened (SMO) inhibitor (squares; n = 13) for 10 days. Data presented as percentages of meca-32–positive areas over analyzed tumor areas for each tumor (Mann-Whitney P = .54; scale bar, 200 um). (B) Quantitative analysis of stromal content by trichromatic stain in pancreatic tumors of KPP mice treated with vehicle or SMO inhibitor for 10 days. Data presented as percentages of positive stain areas over analyzed tissue areas (Mann-Whitney P = .29). (C) Mass spectromic quantitation of intratumoral concentration of 2′,2′-difluorodeoxycytidine triphosphate (dFdCTP; active form of gemcitabine) from each tumor after treatment for 10 consecutive days with vehicle or SMO inhibitor and gemcitabine 50 mg/kg 30 minutes before tumor collection (Mann-Whitney P = 1.000). (D) Ratios of 2′,2′-difluoro 2′-deoxycytidine (dFdC) to diflurodeoxyuridine (dFdU) in pancreatic tumors from each tumor (Mann-Whitney P = .48). NS, not significant.
Fig 5.
Fig 5.
Smoothened (SMO) inhibitor does not affect tumor progression or overall survival in Kras LSL-G12D; p16/p19 fl/fl; Pdx1-Cre (KPP) mice. (A) Individual tumor growth rates plotted by from serial ultrasound images as volumes depicted longitudinally by animal within each regimen. (B) Antilogged values of slopes in each longitudinal plot are graphed, and average tumor burden fold changes per day in each study group of KPP mice are shown, with approximate 95% CIs (vehicle v SMO inhibitor, P = .86; vehicle v combination, P = .0156; gemcitabine v combination, P = .18) (C) Kaplan-Meier plots of KPP mice treated with vehicle (blue, n = 15; median, 1.9 weeks), SMO inhibitor (gold, n = 16; median, 1.2 weeks), gemcitabine (gray, n = 14; median, 3.8 weeks), and gemcitabine plus SMO inhibitor combination (red, n = 12; median, 3.4 weeks; gemcitabine v vehicle, P = .0059; combination v vehicle, P = .0179; gemcitabine v combination, P = .10 [all P values from log-rank test]). HhAntag, hedgehog pathway antagonism.

References

    1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11–30.
    1. Burris HA, 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 1997;15:2403–2413.
    1. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825.
    1. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703.
    1. Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: Phase III trial of the Cancer and Leukemia Group B (CALGB 80303) J Clin Oncol. 2010;28:3617–3622.
    1. Van Cutsem E, Vervenne WL, Bennouna J, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol. 2009;27:2231–2237.
    1. Philip PA, Benedetti J, Corless CL, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group–directed intergroup trial S0205. J Clin Oncol. 2010;28:3605–3610.
    1. Kindler HL, Ioka T, Richel DJ, et al. Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: A double-blind randomised phase 3 study. Lancet Oncol. 2011;12:256–262.
    1. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–1966.
    1. Robarge KD, Brunton SA, Castanedo GM, et al. GDC-0449: A potent inhibitor of the hedgehog pathway. Bioorg Med Chem Lett. 2009;19:5576–5581.
    1. Wong H, Chen JZ, Chou B, et al. Preclinical assessment of the absorption, distribution, metabolism and excretion of GDC-0449 (2-chloro-N-(4-chloro-3-(pyridin-2-yl)phenyl)-4-(methylsulfonyl)benzamide), an orally bioavailable systemic hedgehog signalling pathway inhibitor. Xenobiotica. 2009;39:850–861.
    1. Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009;361:1164–1172.
    1. Rudin CM, Hann CL, Laterra J, et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med. 2009;361:1173–1178.
    1. Catenacci DV. Next-generation clinical trials: Novel strategies to address the challenge of tumor molecular heterogeneity. Mol Oncol. 2015;9:967–996.
    1. LoRusso PM, Rudin CM, Reddy JC, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clinical Cancer Res. 2011;17:2502–2511.
    1. Jimeno A, Weiss GJ, Miller WH, Jr, et al. Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clinical Cancer Res. 2013;19:2766–2774.
    1. Hidalgo M, Maitra A. The hedgehog pathway and pancreatic cancer. N Engl J Med. 2009;361:2094–2096.
    1. Morris JP, 4th, Wang SC, Hebrok M. KRAS, hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nature Rev Cancer. 2010;10:683–695.
    1. Xu FG, Ma QY, Wang Z. Blockade of hedgehog signaling pathway as a therapeutic strategy for pancreatic cancer. Cancer Lett. 2009;283:119–124.
    1. Kelleher FC. Hedgehog signaling and therapeutics in pancreatic cancer. Carcinogenesis. 2011;32:445–451.
    1. Sandhiya S, Melvin G, Kumar SS, et al. The dawn of hedgehog inhibitors: Vismodegib. J Pharmacol Pharmacother. 2013;4:4–7.
    1. Thayer SP, di Magliano MP, Heiser PW, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature. 2003;425:851–856.
    1. Prasad NB, Biankin AV, Fukushima N, et al. Gene expression profiles in pancreatic intraepithelial neoplasia reflect the effects of hedgehog signaling on pancreatic ductal epithelial cells. Cancer Res. 2005;65:1619–1626.
    1. Yauch RL, Gould SE, Scales SJ, et al. A paracrine requirement for hedgehog signalling in cancer. Nature. 2008;455:406–410.
    1. Bailey JM, Swanson BJ, Hamada T, et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clinical Cancer Res. 2008;14:5995–6004.
    1. Tian H, Callahan CA, DuPree KJ, et al. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci U S A. 2009;106:4254–4259.
    1. Theunissen JW, de Sauvage FJ. Paracrine hedgehog signaling in cancer. Cancer Res. 2009;69:6007–6010.
    1. Walter K, Omura N, Hong SM, et al. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clinical Cancer Res. 2010;16:1781–1789.
    1. Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–1461.
    1. Tremblay MR, Lescarbeau A, Grogan MJ, et al. Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926) J Med Chem. 2009;52:4400–4418.
    1. Feldmann G, Fendrich V, McGovern K, et al. An orally bioavailable small-molecule inhibitor of hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer. Mol Cancer Ther. 2008;7:2725–2735.
    1. Feldmann G, Habbe N, Dhara S, et al. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut. 2008;57:1420–1430.
    1. Catenacci D, Bahary N, Horiba N, et al. Final analysis of a phase Ib/Randomized phase II study of Gemcitabine (G) plus Placebo (P) or Vismodegib (V), a hedgehog (Hh) pathway inhibitor, in patients with metastatic pancreatic cancer (PC): A University of Chicago phase II consortium study. J Clin Oncol. 2013;31(suppl 15s):245s. abstr 4012.
    1. Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747.
    1. Lee JJ, Perera RM, Wang H, et al. Stromal response to hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci U S A. 2014;111:E3091–E3100.
    1. Madden JI. Infinity reports update from phase 2 study of saridegib plus gemcitabine in patients with metastatic pancreatic cancer. .
    1. Hingorani SR, Wang L, Multani AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–483.
    1. Romer JT, Kimura H, Magdaleno S, et al. Suppression of the Shh pathway using a small molecule inhibitor eliminates medulloblastoma in Ptc1(+/−)p53(−/−) mice. Cancer Cell. 2004;6:229–240.
    1. Singh M, Lima A, Molina R, et al. Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models. Nat Biotechnol. 2010;28:585–593.
    1. Hruban RH, Adsay NV, Albores-Saavedra J, et al. Pathology of genetically engineered mouse models of pancreatic exocrine cancer: Consensus report and recommendations. Cancer Res. 2006;66:95–106.
    1. Chen W, Tang T, Eastham-Anderson J, et al. Canonical hedgehog signaling augments tumor angiogenesis by induction of VEGF-A in stromal perivascular cells. Proc Natl Acad Sci U S A. 2011;108:9589–9594.
    1. El-Zaatari M, Daignault S, Tessier A, et al. Plasma Shh levels reduced in pancreatic cancer patients. Pancreas. 2012;41:1019–1028.
    1. Kim EJ, Sahai V, Abel EV, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clinical Cancer Res. 2014;20:5937–5945.
    1. Miles KA. Functional computed tomography in oncology. Eur J Cancer. 2002;38:2079–2084.
    1. Abe H, Murakami T, Kubota M, et al. Quantitative tissue blood flow evaluation of pancreatic tumor: Comparison between xenon CT technique and perfusion CT technique based on deconvolution analysis. Radiat Med. 2005;23:364–370.
    1. Kandel S, Kloeters C, Meyer H, et al. Whole-organ perfusion of the pancreas using dynamic volume CT in patients with primary pancreas carcinoma: Acquisition technique, post-processing and initial results. Eur Radiol. 2009;19:2641–2646.
    1. Hattori Y, Gabata T, Matsui O, et al. Enhancement patterns of pancreatic adenocarcinoma on conventional dynamic multi-detector row CT: Correlation with angiogenesis and fibrosis. World J Gastroenterol. 2009;15:3114–3121.
    1. Stoker MG, Shearer M, O'Neill C. Growth inhibition of polyoma-transformed cells by contact with static normal fibroblasts. J Cell Sci. 1966;1:297–310.
    1. Dotto GP, Weinberg RA, Ariza A. Malignant transformation of mouse primary keratinocytes by Harvey sarcoma virus and its modulation by surrounding normal cells. Proc Natl Acad Sci U S A. 1988;85:6389–6393.
    1. Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–348.
    1. Gould SE, Low JA, Marsters JC, Jr, et al. Discovery and preclinical development of vismodegib. Expert Opin Drug Discov. 2014;9:969–984.
    1. Wang Y, Zhou Z, Walsh CT, et al. Selective translocation of intracellular Smoothened to the primary cilium in response to Hedgehog pathway modulation. Proc Natl Acad Sci U S A. 2009;106:2623–2628.
    1. Dijkgraaf GJ, Alicke B, Weinmann L, et al. Small molecule inhibition of GDC-0449 refractory smoothened mutants and downstream mechanisms of drug resistance. Cancer Res. 2011;71:435–444.
    1. Jesus-Acosta A, O'Dwyer PJ, Ramanathan RK, et al. A phase II study of vismodegib, a hedgehog (Hh) pathway inhibitor, combined with gemcitabine and nab-paclitaxel (nab-P) in patients (pts) with untreated metastatic pancreatic ductal adenocarcinoma (PDA) J Clin Oncol. 2014;(suppl 3):32. abstr 257.

Source: PubMed

3
Abonnieren