Genetic delivery of an immunoRNase by an oncolytic adenovirus enhances anticancer activity
Inés Fernández-Ulibarri
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorKatharina Hammer
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorMichaela A.E. Arndt
Department of Translational Medicine, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 560, 69120 Heidelberg, Germany
Search for more papers by this authorJohanna K. Kaufmann
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorDominik Dorer
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorSarah Engelhardt
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorRoland E. Kontermann
Institut für Zellbiologie und Immunologie, Stuttgart University, Allmandring 31, 70569 Stuttgart, Germany
Search for more papers by this authorJochen Hess
Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
Search for more papers by this authorHeike Allgayer
Clinical Cooperation Unit Molecular Oncology of Solid Tumors, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
Department of Experimental Surgery, Medical Faculty Mannheim, University Heidelberg, Theodor-Kutzer-Ufer 1 – 3, 68167 Mannheim, Germany
Search for more papers by this authorJürgen Krauss
Department of Translational Medicine, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 560, 69120 Heidelberg, Germany
Search for more papers by this authorCorresponding Author
Dirk M. Nettelbeck
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Correspondence to: Dirk M. Nettelbeck, Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany, Tel.: +49 (0) 6221 424450, Fax: +49 (0) 6221 421659, E-mail: [email protected]Search for more papers by this authorInés Fernández-Ulibarri
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorKatharina Hammer
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorMichaela A.E. Arndt
Department of Translational Medicine, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 560, 69120 Heidelberg, Germany
Search for more papers by this authorJohanna K. Kaufmann
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorDominik Dorer
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorSarah Engelhardt
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Search for more papers by this authorRoland E. Kontermann
Institut für Zellbiologie und Immunologie, Stuttgart University, Allmandring 31, 70569 Stuttgart, Germany
Search for more papers by this authorJochen Hess
Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
Search for more papers by this authorHeike Allgayer
Clinical Cooperation Unit Molecular Oncology of Solid Tumors, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
Department of Experimental Surgery, Medical Faculty Mannheim, University Heidelberg, Theodor-Kutzer-Ufer 1 – 3, 68167 Mannheim, Germany
Search for more papers by this authorJürgen Krauss
Department of Translational Medicine, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 560, 69120 Heidelberg, Germany
Search for more papers by this authorCorresponding Author
Dirk M. Nettelbeck
Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
Correspondence to: Dirk M. Nettelbeck, Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany, Tel.: +49 (0) 6221 424450, Fax: +49 (0) 6221 421659, E-mail: [email protected]Search for more papers by this authorAbstract
Antibody therapy of solid cancers is well established, but suffers from unsatisfactory tumor penetration of large immunoglobulins or from low serum retention of antibody fragments. Oncolytic viruses are in advanced clinical development showing excellent safety, but suboptimal potency due to limited virus spread within tumors. Here, by developing an immunoRNase-encoding oncolytic adenovirus, we combine viral oncolysis with intratumoral genetic delivery of a small antibody-fusion protein for targeted bystander killing of tumor cells (viro-antibody therapy). Specifically, we explore genetic delivery of a small immunoRNase consisting of an EGFR-binding scFv antibody fragment fused to the RNase Onconase (ONCEGFR) that induces tumor cell death by RNA degradation after cellular internalization. Onconase is a frog RNase that combines lack of immunogenicity and excellent safety in patients with high tumor killing potency due to its resistance to the human cytosolic RNase inhibitor. We show that ONCEGFR expression by oncolytic adenoviruses is feasible with an optimized, replication-dependent gene expression strategy. Virus-encoded ONCEGFR induces potent and EGFR-dependent bystander killing of tumor cells. Importantly, the ONCEGFR-encoding oncolytic adenovirus showed dramatically increased cytotoxicity specifically to EGFR-positive tumor cells in vitro and significantly enhanced therapeutic activity in a mouse xenograft tumor model. The latter demonstrates that ONCEGFR is expressed at levels sufficient to trigger tumor cell killing in vivo. The established ONCEGFR-encoding oncolytic adenovirus represents a novel agent for treatment of EGFR-positive tumors. This viro-antibody therapy platform can be further developed for targeted/personalized cancer therapy by exploiting antibody diversity to target further established or emerging tumor markers or combinations thereof.
Abstract
What's new?
Antibody therapy of solid cancers is well established, but suffers from unsatisfactory tumor penetration of large immunoglobulins or from low serum retention of antibody fragments. Oncolytic viruses are emerging anti-cancer agents that show suboptimal potency due to limited virus spread within tumors. This study establishes an oncolytic virus that genetically delivers a small antibody-based biotherapeutic—an immunoRNase—to tumors. The combined approach addresses barriers to both viral spread and antibody penetration through the expression of small antibody-derivatives inside the tumor that leads to targeted bystander killing and enhanced therapy. Such viro-antibody therapy platform offers interesting opportunities for personalized cancer therapy or targeting of heterologous tumors.
Supporting Information
Additional Supporting Information may be found in the online version of this article.
Filename | Description |
---|---|
ijc29258-sup-0001-suppinfofig1.ppt115 KB |
Supplementary Information |
ijc29258-sup-0002-suppinfofig2.ppt136.5 KB |
Supplementary Information |
ijc29258-sup-0003-suppinfoleg1.docx25.3 KB |
Supplementary Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1 Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30: 658–70.
- 2 Cattaneo R, Miest T, Shashkova EV, Barry MA. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nat Rev Microbiol 2008; 6: 529–40.
- 3 Kaufmann JK, Nettelbeck DM. Virus chimeras for gene therapy, vaccination, and oncolysis: adenoviruses and beyond. Trends Mol Med 2012; 18: 365–76.
- 4 Schrama D, Reisfeld RA, Becker JC. Antibody targeted drugs as cancer therapeutics. Nat Rev Drug Discov 2006; 5: 147–59.
- 5 Batra SK, Jain M, Wittel UA, et al. Pharmacokinetics and biodistribution of genetically engineered antibodies. Curr Opin Biotechnol 2002; 13: 603–8.
- 6 Beckman RA, Weiner LM, Davis HM. Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors. Cancer 2007; 109: 170–9.
- 7 Thurber GM, Schmidt MM, Wittrup KD. Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance. Adv Drug Deliv Rev 2008; 60: 1421–34.
- 8 Rybak SM, Arndt MA, Schirrmann T, et al. Ribonucleases and immunoRNases as anticancer drugs. Curr Pharm Des 2009; 15: 2665–75.
- 9 Ardelt W, Ardelt B, Darzynkiewicz Z. Ribonucleases as potential modalities in anticancer therapy. Eur J Pharmacol 2009; 625: 181–9.
- 10 Costanzi J, Sidransky D, Navon A, Goldsweig H. Ribonucleases as a novel pro-apoptotic anticancer strategy: review of the preclinical and clinical data for ranpirnase. Cancer Invest 2005; 23: 643–50.
- 11 Newton DL, Hansen HJ, Mikulski SM, et al. Potent and specific antitumor effects of an anti-CD22-targeted cytotoxic ribonuclease: potential for the treatment of non-Hodgkin lymphoma. Blood 2001; 97: 528–35.
- 12 Newton D, Pearson J, Xue Y, et al. Anti-tumor ribonuclease, combined with or conjugated to monoclonal antibody MRK16, overcomes multidrug resistance to vincristine in vitro and in vivo. Int J Oncol 1996; 8: 1095–104.
- 13 Chang CH, Sapra P, Vanama SS, et al. Effective therapy of human lymphoma xenografts with a novel recombinant ribonuclease/anti-CD74 humanized IgG4 antibody immunotoxin. Blood 2005; 106: 4308–14.
- 14 Chang CH, Gupta P, Michel R, et al. Ranpirnase (frog RNase) targeted with a humanized, internalizing, anti-Trop-2 antibody has potent cytotoxicity against diverse epithelial cancer cells. Mol Cancer Ther 2010; 9: 2276–86.
- 15 Rybak SM. Antibody-onconase conjugates: cytotoxicity and intracellular routing. Curr Pharm Biotechnol 2008; 9: 226–30.
- 16 Schirrmann T, Krauss J, Arndt MA, et al. Targeted therapeutic RNases (ImmunoRNases). Expert Opin Biol Ther 2009; 9: 79–95.
- 17 Kreitman RJ. Toxin-labeled monoclonal antibodies. Curr Pharm Biotechnol 2001; 2: 313–25.
- 18 Kreitman RJ, Squires DR, Stetler-Stevenson M, et al. Phase I trial of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in patients with B-cell malignancies. J Clin Oncol 2005; 23: 6719–29.
- 19 Messmann RA, Vitetta ES, Headlee D, et al. A phase I study of combination therapy with immunotoxins IgG-HD37-deglycosylated ricin A chain (dgA) and IgG-RFB4-dgA (Combotox) in patients with refractory CD19(+), CD22(+) B cell lymphoma. Clin Cancer Res 2000; 6: 1302–13.
- 20 Boix E, Wu Y, Vasandani VM, et al. Role of the N terminus in RNase A homologues: differences in catalytic activity, ribonuclease inhibitor interaction and cytotoxicity. J Mol Biol 1996; 257: 992–1007.
- 21 Kim BM, Kim H, Raines RT, Lee Y. Glycosylation of onconase increases its conformational stability and toxicity for cancer cells. Biochem Biophys Res Commun 2004; 315: 976–83.
- 22 Siegemund M, Pollak N, Seifert O, et al. Superior antitumoral activity of dimerized targeted single-chain TRAIL fusion proteins under retention of tumor selectivity. Cell Death Dis 2012; 3: e295.
- 23 Goldstein NI, Prewett M, Zuklys K, et al. Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model. Clin Cancer Res 1995; 1: 1311–8.
- 24 He X, Goldsmith CM, Marmary Y, et al. Systemic action of human growth hormone following adenovirus-mediated gene transfer to rat submandibular glands. Gene Ther 1998; 5: 537–41.
- 25 Fueyo J, Gomez-Manzano C, Alemany R, et al. A mutant oncolytic adenovirus targeting the Rb pathway produces anti-glioma effect in vivo. Oncogene 2000; 19: 2–12.
- 26 Rivera AA, Davydova J, Schierer S, et al. Combining high selectivity of replication with fiber chimerism for effective adenoviral oncolysis of CAR-negative melanoma cells. Gene Ther 2004; 11: 1694–702.
- 27 Quirin C, Rohmer S, Fernandez-Ulibarri I, et al. Selectivity and efficiency of late transgene expression by transcriptionally targeted oncolytic adenoviruses are dependent on the transgene insertion strategy. Hum Gene Ther 2011; 22: 389–404.
- 28 Heise C, Hermiston T, Johnson L, et al. An adenovirus E1A mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nat Med 2000; 6: 1134–9.
- 29 Kawakami Y, Li H, Lam JT, et al. Substitution of the adenovirus serotype 5 knob with a serotype 3 knob enhances multiple steps in virus replication. Cancer Res 2003; 63: 1262–9.
- 30 Kim KH, Ryan MJ, Estep JE, et al. A new generation of serotype chimeric infectivity-enhanced conditionally replicative adenovirals: the safety profile of ad5/3-Delta24 in advance of a phase I clinical trial in ovarian cancer patients. Hum Gene Ther 2011; 22: 821–8.
- 31 Kanerva A, Nokisalmi P, Diaconu I, et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF-coding oncolytic adenovirus. Clin Cancer Res 2013; 19: 2734–44.
- 32 Cerullo V, Pesonen S, Diaconu I, et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res 2010; 70: 4297–309.
- 33 Pesonen S, Diaconu I, Cerullo V, et al. Integrin targeted oncolytic adenoviruses Ad5-D24-RGD and Ad5-RGD-D24-GMCSF for treatment of patients with advanced chemotherapy refractory solid tumors. Int J Cancer 2012; 130: 1937–47.
- 34 Koski A, Kangasniemi L, Escutenaire S, et al. Treatment of cancer patients with a serotype 5/3 chimeric oncolytic adenovirus expressing GMCSF. Mol Ther 2010; 18: 1874–84.
- 35 Kim KH, Dmitriev IP, Saddekni S, et al. A phase I clinical trial of Ad5/3-Delta24, a novel serotype-chimeric, infectivity-enhanced, conditionally-replicative adenovirus (CRAd), in patients with recurrent ovarian cancer. Gynecol Oncol 2013; 130: 518–24.
- 36 Liu X, Wu J, Zhang S, Li C, Huang Q. Novel strategies to augment genetically delivered immunotoxin molecular therapy for cancer therapy. Cancer Gene Ther 2009; 16: 861–72.
- 37 Liu X, Li J, Tian Y, et al. Enhanced pancreatic cancer gene therapy by combination of adenoviral vector expressing c-erb-B2 (Her-2/neu)-targeted immunotoxin with a replication-competent adenovirus or etoposide. Hum Gene Ther 2010; 21: 157–70.
- 38 Choi IK, Strauss R, Richter M, et al. Strategies to increase drug penetration in solid tumors. Front Oncol 2013; 3: 193.
- 39 Taylor TE, Furnari FB, Cavenee WK. Targeting EGFR for treatment of glioblastoma: molecular basis to overcome resistance. Curr Cancer Drug Targets 2012; 12: 197–209.
- 40 Fecker LF, Schmude M, Jost S, et al. Efficient and selective tumor cell lysis and induction of apoptosis in melanoma cells by a conditional replication-competent CD95L adenovirus. Exp Dermatol 2010; 19: e56–66.
- 41 Ketzer P, Haas SF, Engelhardt S, et al. Synthetic riboswitches for external regulation of genes transferred by replication-deficient and oncolytic adenoviruses. Nucleic Acids Res 2012; 40: e167.
- 42 Chen H, Sampath P, Hou W, Thorne SH. Regulating cytokine function enhances safety and activity of genetic cancer therapies. Mol Ther 2013; 21: 167–74.
- 43 Rohmer S, Mainka A, Knippertz I, et al. Insulated hsp70B' promoter: stringent heat-inducible activity in replication-deficient, but not replication-competent adenoviruses. J Gene Med 2008; 10: 340–54.
- 44 Zielinski R, Lyakhov I, Jacobs A, et al. Affitoxin-a novel recombinant, HER2-specific, anticancer agent for targeted therapy of HER2-positive tumors. J Immunother 2009; 32: 817–25.
- 45 Behdani M, Zeinali S, Karimipour M, et al. Development of VEGFR2-specific Nanobody Pseudomonas exotoxin A conjugated to provide efficient inhibition of tumor cell growth. N Biotechnol 2013; 30: 205–9.
- 46 Zahnd C, Kawe M, Stumpp MT, et al. Efficient tumor targeting with high-affinity designed ankyrin repeat proteins: effects of affinity and molecular size. Cancer Res 2010; 70: 1595–605.
- 47 Rudnick SI, Lou J, Shaller CC, et al. Influence of affinity and antigen internalization on the uptake and penetration of Anti-HER2 antibodies in solid tumors. Cancer Res 2011; 71: 2250–9.
- 48 Nakase I, Kobayashi S, Futaki S. Endosome-disruptive peptides for improving cytosolic delivery of bioactive macromolecules. Biopolymers 2010; 94: 763–70.
- 49 Arndt MA, Krauss J, Vu BK, et al. A dimeric angiogenin immunofusion protein mediates selective toxicity toward CD22+ tumor cells. J Immunother 2005; 28: 245–51.
- 50 Krauss J, Arndt MA, Vu BK, et al. Targeting malignant B-cell lymphoma with a humanized anti-CD22 scFv-angiogenin immunoenzyme. Br J Haematol 2005; 128: 602–9.
- 51 Menzel C, Schirrmann T, Konthur Z, et al. Human antibody RNase fusion protein targeting CD30+ lymphomas. Blood 2008; 111: 3830–7.