A Phase I-II Study Using Rexin-G Tumor-Targeted Retrovector Encoding a Dominant-Negative Cyclin G1 Inhibitor for Advanced Pancreatic Cancer

Sant P Chawla, Howard Bruckner, Michael A Morse, Nupur Assudani, Frederick L Hall, Erlinda M Gordon, Sant P Chawla, Howard Bruckner, Michael A Morse, Nupur Assudani, Frederick L Hall, Erlinda M Gordon

Abstract

Rexin-G is a replication-incompetent retroviral vector displaying a cryptic SIG-binding peptide for targeting abnormal Signature (SIG) proteins in tumors and encoding a dominant-negative human cyclin G1 construct. Herein we report on the safety and antitumor activity of escalating doses of Rexin-G in gemcitabine-refractory pancreatic adenocarcinoma, with one 10-year survivor. For the safety analysis (n = 20), treatment-related grade 1 adverse events included fatigue (n = 6), chills (n = 2), and headache (n = 1), with no organ damage and no DLT. No patient tested positive for vector-neutralizing antibodies, antibodies to gp70, replication-competent retrovirus (RCR), or vector integration into genomic DNA of peripheral blood lymphocytes (PBLs). For the efficacy analysis (n = 15), one patient achieved a complete response (CR), two patients had a partial response (PR), and 12 had stable disease (SD). Median progression-free survival (PFS) was 2.7, 4.0, and 5.6 months at doses 0-I, II, and III, respectively. Median overall survival (OS) and 1-year OS rate at dose 0-I were 4.3 months and 0%, and at dose II-III they were 9.2 months and 33.3%. To date, one patient is still alive with no evidence of cancer 10 years after the start of Rexin-G treatment. Taken together, these data suggest that Rexin-G, the first targeted gene delivery system, is uniquely safe and exhibits significant antitumor activity, for which the FDA granted fast-track designation.

Keywords: 10-year cancer-free survivor; CCNG1; cell cycle control; cyclin G1 inhibitor; gene therapy; pancreas adenocarcinoma; retrovector; targeted gene delivery; tumor targeting.

Figures

Figure 1
Figure 1
Graphic Illustration of Rexin-G Vector The Rexin-G vector displaying a SIG-targeting peptide (A), for binding to Signature (SIG) proteins in the tumor microenvironment (TME) (B), and encoding a dominant-negative human cyclin G1 inhibitor gene (C). Injected intravenously, Rexin-G nanoparticles seek out and bind to abnormal SIG proteins in the TME, which augments effective vector concentration in tumors.
Figure 2
Figure 2
Kaplan-Meier Plot of Progression-free Survival in Rexin-G-Treated Gemcitabine-Resistant PDAC Progression-free survival data for the modified intent-to-treat population are displayed. The proportion of patients surviving progression-free are plotted on the vertical axis as a function of time from the beginning of treatment, plotted on the horizontal axis.
Figure 3
Figure 3
Tumor Regression during Treatment with Rexin-G Patients 12 (A) and 16 (B). Percentage change in tumor size (sum of longest diameter [SLD]) is plotted on the vertical axis, as a function of time from the beginning of Rexin-G treatment, plotted on the horizontal axis.
Figure 4
Figure 4
Tumor Regression and CA-19.9 Levels during Treatment with Rexin-G in Patient 18 (A) Percentage changes in tumor size (longest diameter [LD]) of metastatic hepatic and lymph node sub-peritoneal lesions are individually plotted on the vertical axis, as a function of time from the beginning of Rexin-G treatment, plotted on the horizontal axis. (B) Serum levels of tumor marker CA-19.9 (U/mL) are plotted on the veritical axis, as a function of time from the beginning of Rexin-G treatment, plotted on the horizontal axis.
Figure 5
Figure 5
Kaplan-Meier Plot of Overall Survival of PDAC Patients following Rexin-G Treatment at Escalating Dose Levels Analysis of overall survival in the ITT population suggests a dose-response relationship between overall survival and Rexin-G dosage (p = 0.03). The proportion of patients surviving are plotted on the vertical axis as a function of time from beginning of treatment, plotted on the horizontal axis.

References

    1. Rahib L., Smith B.D., Aizenberg R., Rosenzweig A.B., Fleshman J.M., Matrisian L.M. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–2921.
    1. American Cancer Society (2018). Cancer facts & figures 2018. .
    1. Conroy T., Desseigne F., Ychou M., Bouché O., Guimbaud R., Bécouarn Y., Adenis A., Raoul J.L., Gourgou-Bourgade S., de la Fouchardière C., Groupe Tumeurs Digestives of Unicancer. PRODIGE Intergroup FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011;364:1817–1825.
    1. Von Hoff D.D., Ervin T., Arena F.P., Chiorean E.G., Infante J., Moore M., Seay T., Tjulandin S.A., Ma W.W., Saleh M.N. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013;369:1691–1703.
    1. Wang-Gillam A., Li C.P., Bodoky G., Dean A., Shan Y.S., Jameson G., Macarulla T., Lee K.H., Cunningham D., Blanc J.F., NAPOLI-1 Study Group Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387:545–557.
    1. Moore M.J., Goldstein D., Hamm J., Figer A., Hecht J.R., Gallinger S., Au H.J., Murawa P., Walde D., Wolff R.A., National Cancer Institute of Canada Clinical Trials Group 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. Liu Y., Elsawa S.F., Almada L.L. Primers on molecular pathways - cycling toward pancreatic cancer. Pancreatology. 2010;10:6–13.
    1. Matera R., Saif M.W. New therapeutic directions for advanced pancreatic cancer: cell cycle inhibitors, stromal modifiers and conjugated therapies. Expert Opin. Emerg. Drugs. 2017;22:223–233.
    1. Waehler R., Russell S.J., Curiel D.T. Engineering targeted viral vectors for gene therapy. Nat. Rev. Genet. 2007;8:573–587.
    1. Hall F.L., Liu L., Zhu N.L., Stapfer M., Anderson W.F., Beart R.W., Gordon E.M. Molecular engineering of matrix-targeted retroviral vectors incorporating a surveillance function inherent in von Willebrand factor. Hum. Gene Ther. 2000;11:983–993.
    1. Xu F., Prescott M.F., Liu P.X., Chen Z.H., Liau G., Gordon E.M., Hall F.L. Long term inhibition of neointima formation in balloon-injured rat arteries by intraluminal instillation of a matrix-targeted retroviral vector bearing a cytocidal mutant cyclin G1 construct. Int. J. Mol. Med. 2001;8:19–30.
    1. Gordon E.M., Hall F.L. Rexin-G, a targeted genetic medicine for cancer. Expert Opin. Biol. Ther. 2010;10:819–832.
    1. Gordon E.M., Liu P.X., Chen Z.H., Liu L., Whitley M.D., Gee C., Groshen S., Hinton D.R., Beart R.W., Hall F.L. Inhibition of metastatic tumor growth in nude mice by portal vein infusions of matrix-targeted retroviral vectors bearing a cytocidal cyclin G1 construct. Cancer Res. 2000;60:3343–3347.
    1. Gordon E.M., Chen Z.H., Liu L., Whitley M., Liu L., Wei D., Groshen S., Hinton D.R., Anderson W.F., Beart R.W., Jr., Hall F.L. Systemic administration of a matrix-targeted retroviral vector is efficacious for cancer gene therapy in mice. Hum. Gene Ther. 2001;12:193–204.
    1. Gordon E.M., Cornelio G.H., Lorenzo C.C., 3rd, Levy J.P., Reed R.A., Liu L., Hall F.L. First clinical experience using a ‘pathotropic’ injectable retroviral vector (Rexin-G) as intervention for stage IV pancreatic cancer. Int. J. Oncol. 2004;24:177–185.
    1. Gordon E.M., Lopez F.F., Cornelio G.H., Lorenzo C.C., 3rd, Levy J.P., Reed R.A., Liu L., Bruckner H.W., Hall F.L. Pathotropic nanoparticles for cancer gene therapy Rexin-G IV: three-year clinical experience. Int. J. Oncol. 2006;29:1053–1064.
    1. Galanis E., Carlson S.K., Foster N.R., Lowe V., Quevedo F., McWilliams R.R., Grothey A., Jatoi A., Alberts S.R., Rubin J. Phase I trial of a pathotropic retroviral vector expressing a cytocidal cyclin G1 construct (Rexin-G) in patients with advanced pancreatic cancer. Mol. Ther. 2008;16:979–984.
    1. Chawla S.P., Chua V.S., Fernandez L., Quon D., Saralou A., Blackwelder W.C., Hall F.L., Gordon E.M. Phase I/II and phase II studies of targeted gene delivery in vivo: intravenous Rexin-G for chemotherapy-resistant sarcoma and osteosarcoma. Mol. Ther. 2009;17:1651–1657.
    1. Chawla S.P., Chua V.S., Fernandez L., Quon D., Blackwelder W.C., Gordon E.M., Hall F.L. Advanced phase I/II studies of targeted gene delivery in vivo: intravenous Rexin-G for gemcitabine-resistant metastatic pancreatic cancer. Mol. Ther. 2010;18:435–441.
    1. Choi H., Charnsangavej C., Faria S.C., Macapinlac H.A., Burgess M.A., Patel S.R., Chen L.L., Podoloff D.A., Benjamin R.S. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J. Clin. Oncol. 2007;25:1753–1759.
    1. NCI Common terminology criteria for adverse events v3.0. 2006.
    1. Dy P.S.G., Chawla S.P., Hall F.L., Gordon E.M. Immune cell trafficking in the tumor microenvironment of human cyclin G1 (CCNG1) inhibitor-treated tumors. Br. J. Cancer Res. 2018;1:202–207.
    1. Burris H.A., 3rd, Moore M.J., Andersen J., Green M.R., Rothenberg M.L., Modiano M.R., Cripps M.C., Portenoy R.K., Storniolo A.M., Tarassoff P. 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. Berlin J.D., Catalano P., Thomas J.P., Kugler J.W., Haller D.G., Benson A.B., 3rd Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J. Clin. Oncol. 2002;20:3270–3275.
    1. Rocha Lima C.M., Green M.R., Rotche R., Miller W.H., Jr., Jeffrey G.M., Cisar L.A., Morganti A., Orlando N., Gruia G., Miller L.L. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J. Clin. Oncol. 2004;22:3776–3783.
    1. Heinemann V., Quietzsch D., Gieseler F., Gonnermann M., Schönekäs H., Rost A., Neuhaus H., Haag C., Clemens M., Heinrich B. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J. Clin. Oncol. 2006;24:3946–3952.
    1. Colucci G., Giuliani F., Gebbia V., Biglietto M., Rabitti P., Uomo G., Cigolari S., Testa A., Maiello E., Lopez M. Gemcitabine alone or with cisplatin for the treatment of patients with locally advanced and/or metastatic pancreatic carcinoma: a prospective, randomized phase III study of the Gruppo Oncologia dell’Italia Meridionale. Cancer. 2002;94:902–910.
    1. Louvet C., Labianca R., Hammel P., Lledo G., Zampino M.G., André T., Zaniboni A., Ducreux M., Aitini E., Taïeb J., GERCOR. GISCAD Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J. Clin. Oncol. 2005;23:3509–3516.
    1. Poplin E., Feng Y., Berlin J., Rothenberg M.L., Hochster H., Mitchell E., Alberts S., O’Dwyer P., Haller D., Catalano P. Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: a trial of the Eastern Cooperative Oncology Group. J. Clin. Oncol. 2009;27:3778–3785.
    1. Herrmann R., Bodoky G., Ruhstaller T., Glimelius B., Bajetta E., Schüller J., Saletti P., Bauer J., Figer A., Pestalozzi B., Swiss Group for Clinical Cancer Research. Central European Cooperative Oncology Group Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J. Clin. Oncol. 2007;25:2212–2217.
    1. Burris H., 3rd, Rocha-Lima C. New therapeutic directions for advanced pancreatic cancer: targeting the epidermal growth factor and vascular endothelial growth factor pathways. Oncologist. 2008;13:289–298.
    1. Senderowicz A.M., Johnson J.R., Sridhara R., Zimmerman P., Justice R., Pazdur R. Erlotinib/gemcitabine for first-line treatment of locally advanced or metastatic adenocarcinoma of the pancreas. Oncology (Williston Park) 2007;21:1696–1706.
    1. Gordon, E.M., and Hall, F.L. Cyclin G1 inhibitors and related methods of treating cancer. US patent WO/2018/144863, filed February 2, 2018 and granted September 8, 2018.
    1. Schuetz T., Kaufman H.L., Marshall J.L., Safran H. Extended survival in second-line pancreatic cancer after therapeutic vaccination. J. Clin. Oncol. 2005;23:2576.
    1. Petrulio C.A., Kaufman H.L. Development of the PANVAC-VF vaccine for pancreatic cancer. Expert Rev. Vaccines. 2006;5:9–19.
    1. FDA News (2018). Therion reports results of PANVAC-VF trial. .
    1. Kim S., Federman N., Gordon E.M., Hall F.L., Chawla S.P. Rexin-G®, a tumor-targeted retrovector for malignant peripheral nerve sheath tumor: A case report. Mol. Clin. Oncol. 2017;6:861–865.
    1. Gordon E.M., Ravicz J.R., Liu S., Chawla S.P., Hall F.L. Cell cycle checkpoint control: The cyclin G1/Mdm2/p53 axis emerges as a strategic target for broad-spectrum cancer gene therapy - A review of molecular mechanisms for oncologists. Mol. Clin. Oncol. 2018;9:115–134.
    1. Ravicz J., Liu S., Andrali S.S., Reddy S.K., Sellappan S., Leong B., Chawla S.P., Hall F.L., Gordon E.M. Differential expression of human cyclin G1 (CCNG1) in cancer: A novel biomarker in development for CCNG1 inhibitor therapy. J. Clin. Oncol. 2018;36:e24315.
    1. Hall F.L., Levy J.P., Reed R.A., Petchpud W.N., Chua V.S., Chawla S.P., Gordon E.M. Pathotropic targeting advances clinical oncology: tumor-targeted localization of therapeutic gene delivery. Oncol. Rep. 2010;24:829–833.
    1. FDA (2006). Guidance for industry: supplemental guidance on testing for replication competent retrovirus in retroviral vector based gene therapy products and during follow-up of patients in clinical trials using retroviral vectors. .
    1. Therasse P., Arbuck S.G., Eisenhauer E.A., Wanders J., Kaplan R.S., Rubinstein L., Verweij J., Van Glabbeke M., van Oosterom A.T., Christian M.C., Gwyther S.G. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J. Natl. Cancer Inst. 2000;92:205–216.
    1. Young H., Baum R., Cremerius U., Herholz K., Hoekstra O., Lammertsma A.A., Pruim J., Price P., European Organization for Research and Treatment of Cancer (EORTC) PET Study Group Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. Eur. J. Cancer. 1999;35:1773–1782.

Source: PubMed

3
Abonneren