VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects

Miriam Bazan-Peregrino, Rocio Garcia-Carbonero, Berta Laquente, Rafael Álvarez, Ana Mato-Berciano, Marta Gimenez-Alejandre, Sara Morgado, Alba Rodríguez-García, Maria V Maliandi, M Carmen Riesco, Rafael Moreno, Mireia M Ginestà, Mercedes Perez-Carreras, Joan B Gornals, Susana Prados, Sofía Perea, Gabriel Capella, Ramon Alemany, Ramon Salazar, Emma Blasi, Carmen Blasco, Manel Cascallo, Manuel Hidalgo, Miriam Bazan-Peregrino, Rocio Garcia-Carbonero, Berta Laquente, Rafael Álvarez, Ana Mato-Berciano, Marta Gimenez-Alejandre, Sara Morgado, Alba Rodríguez-García, Maria V Maliandi, M Carmen Riesco, Rafael Moreno, Mireia M Ginestà, Mercedes Perez-Carreras, Joan B Gornals, Susana Prados, Sofía Perea, Gabriel Capella, Ramon Alemany, Ramon Salazar, Emma Blasi, Carmen Blasco, Manel Cascallo, Manuel Hidalgo

Abstract

Background: Pancreatic ductal adenocarcinoma (PDAC) is characterized by dense desmoplastic stroma that limits the delivery of anticancer agents. VCN-01 is an oncolytic adenovirus designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase. Here, we evaluated the mechanism of action of VCN-01 in preclinical models and in patients with pancreatic cancer.

Methods: VCN-01 replication and antitumor efficacy were evaluated alone and in combination with standard chemotherapy in immunodeficient and immunocompetent preclinical models using intravenous or intratumoral administration. Hyaluronidase activity was evaluated by histochemical staining and by measuring drug delivery into tumors. In a proof-of-concept clinical trial, VCN-01 was administered intratumorally to patients with PDAC at doses up to 1×1011 viral particles in combination with chemotherapy. Hyaluronidase expression was measured in serum by an ELISA and its activity within tumors by endoscopic ultrasound elastography.

Results: VCN-01 replicated in PDAC models and exerted antitumor effects which were improved when combined with chemotherapy. Hyaluronidase expression by VCN-01 degraded tumor stroma and facilitated delivery of a variety of therapeutic agents such as chemotherapy and therapeutic antibodies. Clinically, treatment was generally well-tolerated and resulted in disease stabilization of injected lesions. VCN-01 was detected in blood as secondary peaks and in post-treatment tumor biopsies, indicating virus replication. Patients had increasing levels of hyaluronidase in sera over time and decreased tumor stiffness, suggesting stromal disruption.

Conclusions: VCN-01 is an oncolytic adenovirus with direct antitumor effects and stromal disruption capabilities, representing a new therapeutic agent for cancers with dense stroma.

Trial registration number: EudraCT number: 2012-005556-42 and NCT02045589.

Keywords: gastrointestinal neoplasms; immunity; investigational; oncolytic viruses; therapies; tumor microenvironment.

Conflict of interest statement

Competing interests: MB-P, AM-B, MG-A, SM, MVM, EB, CB and MC are employees and RAlm and MH are consultants for VCN Biosciences. MC and RAlm are co-inventors of one patent application concerning the expression of hyaluronidase by oncolytic adenoviruses. MC, RAlm and GC have ownership interest in VCN Biosciences. MH is a founder and stock holder of Champions Oncology and Nelum Pharmaceuticals; he is a stock holder of Agenus, Pharmacyte, Highlight Pharmaceuticals, Oncomatrix, Inxmed and BMS; he is Board of Directors at BMS; he has received research support from Erytech, PanCan, TBA alliance and has received honorarium from Agenus, Oncomatrix, InxMed, Khar, Genechem, Cantargia, BMS, and royalties from Myriad and Kahr. RGC has provided scientific advice and/or received honoraria or funding for continuous medical education from AAA, Advanz Pharma, Amgen, Bayer, BMS, HMP, Ipsen, Merck, Mida-tech Pharma, MSD, Novartis, PharmaMar, Pfizer, Pierre Fabre, Roche, Servier and Sanofi, and has received research support from Pfizer, BMS and MSD.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
VCN-01 sensitizes tumors to chemotherapy leading to enhanced antitumor activity. (A) Antitumor activity against subcutaneous HP-1 Syrian hamster pancreatic tumors, implanted in immunocompetent hamsters, was evaluated after three intratumoral administrations of 2.5×1010 vp of VCN-01 per tumor alone (days 0, 9 and 18) or in combination with intraperitoneal (IP) G for seven doses (50 mg/kg, days 0, 3, 6, 9, 12, 15 and 18). n=8–12 tumors/group. (B) Antitumor activity against subcutaneous human NP-9 pancreatic tumors, implanted in immunodeficient mice, was evaluated after single intravenous administration of 4×1010 viral particles (VP) of VCN-01 alone (day 0) or in combination with IP gemcitabine (G) (50 mg/kg, days 0, 3, 6 and 9). n=7–13 tumors/group. (C) Viral E1A expression and hyaluronic acid (HA) degradation indicative of viral replication as determined by immunohistochemistry on serial tumor sections of VCN-01 treated tumors of (B) on day 79. (D) Antitumor activity against subcutaneous human pancreatic NP-18 tumors, implanted in immunodeficient mice, was evaluated after single intravenous administration of VCN-01 (4×1010vp/animal, day 0) with and without IP G with nab-paclitaxel plus gemcitabine (GA) (G 50 mg/kg and a 41.6 mg/kg, days 0, 7 and 14). n=10–12 tumors/group. (E) Tumor volumes for each tumor treated with VCN-01 +GA. For A, B, D, a parametric t-student test was used to determine statistical significance of the different treatments at each time point. (F) Antitumor activity against the orthotopic human TP-11 pancreatic model, implanted in immunodeficient mice, after single intravenous administration of VCN-01 (4×1010 vp/animal, day 0) with and without IP GA (a 41.6 mg/kg & g 50 mg/kg, days 0, 7 and 14) and measured tumor volume at the experiment end (day 28). n=6–13 tumors/group. Mean±SEM is plotted (black lines). One-way ANOVA (Tukey’s multiple comparisons test) was used to determine statistical significance of the different treatments. ANOVA, analysis of variance; GA, gemcitabine with albumin.
Figure 2
Figure 2
VCN-01 enhances extravasation of chemotherapy and monoclonal antibodies into tumors. (A) Single intravenous administration of VCN-01 alone or in combination with chemotherapy significantly increases tumor-uptake of gemcitabine (G) in an orthotopic pancreatic model, TP-11. Animals from the different experimental groups described in figure 1F were administered G IP at a dose of 50 mg/kg 1 hour before sacrifice. The intratumoral content of G in each tumor was measured by high-performance liquid chromatography (HPLC). Box and whiskers plots (mean is shown as +). n=6–13 tumors/group. One-way ANOVA (Tukey’s multiple comparisons test) was employed to determine statistical significance of the different treatments. (B) fluorescence in vivo imaging after intravenous administration of labeled anti-PD-L1 antibody (10 mg/kg). VCN-01 or the hyaluronidase negative virus ICOVIR15K was administered intravenous at a dose of 4×1010 vp/animal to NP-18 tumor-bearing immunodeficient mice on day 0. Labeled anti-PD-L1 (Vivotag 800) was administered intravenous at a dose of 10 mg/kg on day 14 and imaging performed after 48 hours. Upper panel: representative images from fluorescence in tumors show enhanced extravasation of labeled anti-PD-L1 in VCN-01 treated tumors compared with the ICOVIR15K group. Lower panel: quantification of average radiant efficiency at 800 nm in tumor areas is represented in the box and whiskers plot (mean is shown as +). n=6–8 tumors/group. One-way ANOVA (Tukey’s multiple comparisons test) was employed to determine statistical significance of the different treatments. (C) tumor growth curves of the TRAMPC-2 cancer model treated with 1×1010vp/tumor of VCN-01 or phosphate-buffered saline administered intratumorally alone or in combination with five intraperitoneal doses of 200 µg of anti-PD-L1 antibody (days 0, 3, 6, 8 and 10). n=8–9 tumors/group. A two-way ANOVA, followed by a Tukey’s multiple comparisons test, was employed to determine statistical significance of the different treatments for each day. ANOVA, analysis of variance; GA, gemcitabine with albumin.
Figure 3
Figure 3
VCN-01 pharmacokinetics in mice and intratumoral viral replication. (A) Pharmacokinetics and peaks of replication of VCN-01 in blood after single intratumoral administration of 5×108 vp in athymic mice with the human pancreatic tumor NP-18 implanted subcutaneous (SC). Blood samples (20 µL) were obtained at the indicated time points postadministration and viral genomes were measured by qPCR. n=6–7 mice/time point. (B) Left panel: VCN-01 blood levels after administration of a single intravenous dose of 5×1010 vp/animal to immunodeficient mice bearing the SC human pancreatic NP-18 tumor. Blood samples (20 µL) were obtained at 2 and 15 min and at 1, 4, 24, and 48 hours. Post dose and genomes were measured by qPCR. Right panel: VCN-01 viral genomes in blood of immunodeficient mice with the SC NP-18 human pancreatic model after intravenous administration of 4×1010 vp/animal of VCN-01 on day 0. Blood samples (20 µL) were collected on days 2, 10 and 21. Results are expressed as viral genomes per mL of total blood. n=6–7 mice/time point. (C) Viral genomes in tumors and livers were analyzed at the indicated time points after one time intravenous administration of VCN-01 (4×1010 vp/animal) to mice with orthotopically implanted TP-11 human pancreatic tumor model. Animals were sacrificed on day 2 and 28 and VCN-01 genome measured by qPCR. n=6–8 tumors or livers/group/day. A Mann-Whitney U test was employed to analyze statistically significant differences (*p<0.05 vs D2).
Figure 4
Figure 4
Antitumor activity of Intratumor Injections of VCN-01 in patients. Percentage variation in the size of tumor lesions injected with VCN-01 compared with baseline in patients treated with VCN-01 in combination with either gemcitabine (G) or nab-paclitaxel plus gemcitabine (GA). Ech blue line represents the monitored tumor of each patient (eight patients).
Figure 5
Figure 5
VCN-01 is detected in patient blood over time showing viral replication peaks and in tumors 20–28 days after treatment. (A) Blood levels of VCN-01 at the indicated time points as determined by qPCR. Each black line represents a patient (six patients) and the mean of all patients treated with 1×1011 vp + nab-paclitaxel plus gemcitabine (GA) per time point is shown in gray. (B) VCN-01 is detected in all tumors analyzed on days 20–28. Fine-needle aspiration biopsy of the tumor performed at the indicated times (pre-VCN-01 or post-treatment) on each patient and analyzed by qPCR to detect the number of viral genomes (six patients). Each tumor sample is represented by a symbol, performed either on day 0 (negative) or between day 20 and 28 post-VCN-01 it administration. G, gemcitabine.
Figure 6
Figure 6
Hyaluronidase PH20 expression and stroma disruption effects of VCN-01 in patients. (A) PH20 serum levels as determined by ELISA at the indicated time points in patients treated with VCN-01 in combination with nab-paclitaxel plus gemcitabine (GA). Each filled circle represents a serum sample per patient. Analyzed patients were injected at a dose of 1×1011 vp of VCN-01 (six patients). One-way ANOVA (Tukey’s multiple comparisons test) was used to evaluate multiple comparisons between groups. PH20 expression in serum samples at different time points is expressed as pg/mL minus background levels detected for each sample at day 0 and represented in box and whiskers graphs. (B) Changes in strain ratio measured by endoscopic ultrasound (EUS) elastography in five patients prior to cycles 1, 2 and 3. Each line represents an individual patient. ANOVA, analysis of variance.

References

    1. Korc M. Pancreatic cancer-associated stroma production. Am J Surg 2007;194:S84–6. 10.1016/j.amjsurg.2007.05.004
    1. Sun C, Ansari D, Andersson R, et al. . Does gemcitabine-based combination therapy improve the prognosis of unresectable pancreatic cancer? World J Gastroenterol 2012;18:4944–58. 10.3748/wjg.v18.i35.4944
    1. Rasheed ZA, Matsui W, Maitra A. Pathology of pancreatic stroma in PDAC. In: Grippo PJ, Munshi HG, eds. Pancreatic cancer and tumor microenvironment. Trivandrum (India), 2012.
    1. Nielsen MFB, Mortensen MB, Detlefsen S. Key players in pancreatic cancer-stroma interaction: cancer-associated fibroblasts, endothelial and inflammatory cells. World J Gastroenterol 2016;22:2678–700. 10.3748/wjg.v22.i9.2678
    1. Chu GC, Kimmelman AC, Hezel AF, et al. . Stromal biology of pancreatic cancer. J Cell Biochem 2007;101:887–907. 10.1002/jcb.21209
    1. Erkan M, Reiser-Erkan C, Michalski CW, et al. . Tumor microenvironment and progression of pancreatic cancer. Exp Oncol 2010;32:128–31.
    1. Whatcott CJ, Han H, Posner RG, et al. . Targeting the tumor microenvironment in cancer: why hyaluronidase deserves a second look. Cancer Discov 2011;1:291–6. 10.1158/-11-0136
    1. Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer 2004;4:528–39. 10.1038/nrc1391
    1. Bertrand P, Girard N, Delpech B, et al. . Hyaluronan (hyaluronic acid) and hyaluronectin in the extracellular matrix of human breast carcinomas: comparison between invasive and non-invasive areas. Int J Cancer 1992;52:1–6. 10.1002/ijc.2910520102
    1. Seufferlein T, Ducreux M, Hidalgo M, et al. . More than a Gel & Hyaluronic Acid, a Central Component in the Microenvironment of Pancreatic Cancer. Eur Oncol Haematol 2018;14:40–4. 10.17925/EOH.2018.14.1.40
    1. Oettle H, Post S, Neuhaus P, et al. . Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA 2007;297:267–77. 10.1001/jama.297.3.267
    1. Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer 2006;6:583–92. 10.1038/nrc1893
    1. Feig C, Gopinathan A, Neesse A, et al. . The pancreas cancer microenvironment. Clin Cancer Res 2012;18:4266–76. 10.1158/1078-0432.CCR-11-3114
    1. Nakazawa H, Yoshihara S, Kudo D, et al. . 4-Methylumbelliferone, a hyaluronan synthase suppressor, enhances the anticancer activity of gemcitabine in human pancreatic cancer cells. Cancer Chemother Pharmacol 2006;57:165–70. 10.1007/s00280-005-0016-5
    1. Provenzano PP, Cuevas C, Chang AE, et al. . Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012;21:418–29. 10.1016/j.ccr.2012.01.007
    1. Singha NC, Nekoroski T, Zhao C, et al. . Tumor-Associated hyaluronan limits efficacy of monoclonal antibody therapy. Mol Cancer Ther 2015;14:523–32. 10.1158/1535-7163.MCT-14-0580
    1. Hendifar A, Bullock A. Breaking the Barrier—PEGylated RecombinantHuman Hyaluronidase (PEGPH20)—A New Therapeutic Approach to the Treatment of Pancreatic Ductal Adenocarcinoma. Oncol Hematol Rev 2017;13:107–11. 10.17925/OHR.2017.13.02.107
    1. Rodríguez-García A, Giménez-Alejandre M, Rojas JJ, et al. . Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin Cancer Res 2015;21:1406–18. 10.1158/1078-0432.CCR-14-2213
    1. Guedan S, Rojas JJ, Gros A, et al. . Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol Ther 2010;18:1275–83. 10.1038/mt.2010.79
    1. Vives M, Ginestà MM, Gracova K, et al. . Metronomic chemotherapy following the maximum tolerated dose is an effective anti-tumour therapy affecting angiogenesis, tumour dissemination and cancer stem cells. Int J Cancer 2013;133:2464–72. 10.1002/ijc.28259
    1. Janssen J, Schlörer E, Greiner L. EUS elastography of the pancreas: feasibility and pattern description of the normal pancreas, chronic pancreatitis, and focal pancreatic lesions. Gastrointest Endosc 2007;65:971–8. 10.1016/j.gie.2006.12.057
    1. Iglesias-Garcia J, Lindkvist B, Lariño-Noia J, et al. . Endoscopic ultrasound elastography. Endosc Ultrasound 2012;1:8–16. 10.4103/2303-9027.117764
    1. Alvarez R, Musteanu M, Garcia-Garcia E, et al. . Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br J Cancer 2013;109:926–33. 10.1038/bjc.2013.415
    1. Hecht JR, Bedford R, Abbruzzese JL, et al. . A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res 2003;9:555–61.
    1. Liang M, Oncorine LM. Oncorine, the world first oncolytic virus medicine and its update in China. Curr Cancer Drug Targets 2018;18:171–6. 10.2174/1568009618666171129221503
    1. Conry RM, Westbrook B, McKee S, et al. . Talimogene laherparepvec: first in class oncolytic virotherapy. Hum Vaccin Immunother 2018;14:839–46. 10.1080/21645515.2017.1412896
    1. Garcia-Carbonero R, Gil Martín M, Alvarez Gallego R, et al. . Systemic administration of the hyaluronidase-expressing oncolytic adenovirus VCN-01 in patients with advanced or metastatic pancreatic cancer: first-in-human clinical trial. Annals Oncol 2019;30:v271–2. 10.1093/annonc/mdz247.037
    1. Thompson CB, Shepard HM, O'Connor PM, et al. . Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol Cancer Ther 2010;9:3052–64. 10.1158/1535-7163.MCT-10-0470
    1. Jacobetz MA, Chan DS, Neesse A, et al. . Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013;62:112–20. 10.1136/gutjnl-2012-302529
    1. Cascallo M, Alonso MM, Rojas JJ, et al. . Systemic toxicity-efficacy profile of ICOVIR-5, a potent and selective oncolytic adenovirus based on the pRB pathway. Mol Ther 2007;15:1607–15. 10.1038/sj.mt.6300239
    1. de Sostoa J, Fajardo CA, Moreno R, et al. . Targeting the tumor stroma with an oncolytic adenovirus secreting a fibroblast activation protein-targeted bispecific T-cell engager. J Immunother Cancer 2019;7:19. 10.1186/s40425-019-0505-4
    1. Toro Bejarano M, Merchan JR. Targeting tumor vasculature through oncolytic virotherapy: recent advances. Oncolytic Virother 2015;4:169–81. 10.2147/OV.S66045
    1. Bazan-Peregrino M, Sainson RCA, Carlisle RC, et al. . Combining virotherapy and angiotherapy for the treatment of breast cancer. Cancer Gene Ther 2013;20:461–8. 10.1038/cgt.2013.41
    1. Bockorny B, Semenisty V, Macarulla T, et al. . BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the combat trial. Nat Med 2020;26:878–85. 10.1038/s41591-020-0880-x
    1. Royal RE, Levy C, Turner K, et al. . Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010;33:828–33. 10.1097/CJI.0b013e3181eec14c
    1. O'Reilly EM, DY O, Dhani N. Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol 2019.
    1. Pascual-Pasto G, Bazan-Peregrino M, Olaciregui NG, et al. . Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01. Sci Transl Med 2019;11. 10.1126/scitranslmed.aat9321. [Epub ahead of print: 23 01 2019].
    1. Ramanathan RK, McDonough SL, Philip PA, et al. . Phase Ib/II randomized study of Folfirinox plus PEGylated recombinant human hyaluronidase versus Folfirinox alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol 2019;37:1062–9. 10.1200/JCO.18.01295
    1. Tempero MA, Van Cutsem E, Sigal D, et al. . HALO 109-301: A randomized, double-blind, placebo-controlled, phase 3 study of pegvorhyaluronidase alfa (PEGPH20) + nab-paclitaxel/gemcitabine (AG) in patients (pts) with previously untreated hyaluronan (HA)-high metastatic pancreatic ductal adenocarcinoma (mPDA). JCO 2020;38:638–38. 10.1200/JCO.2020.38.4_suppl.638
    1. Rhim AD, Oberstein PE, Thomas DH, Rhim Andrew D, Thomas Dafydd H, et al. . Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014;25:735–47. 10.1016/j.ccr.2014.04.021

Source: PubMed

3
Subscribe