TumorDiagnostik & Therapie 2012; 33(5): 279-283
DOI: 10.1055/s-0032-1313090
Thieme Onkologie aktuell
© Georg Thieme Verlag KG Stuttgart · New York

Oncolytic Viruses to Treat Ovarian Cancer Patients – a Review of Results From Clinical Trials

Onkolytische Viren zur Behandlung von Ovarialkarzinompatientinnen – Eine Übersicht der Ergebnisse klinischer Studien
A. D. Hartkopf
1   Department of Obstetrics and Gynecology, University of Tübingen, Tübingen
,
T. Fehm
1   Department of Obstetrics and Gynecology, University of Tübingen, Tübingen
,
M. Wallwiener
2   Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg
,
U. Lauer
3   Department of Gastroenterology and Hepatology, University of Tübingen, Tübingen
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Publikationsdatum:
23. August 2012 (online)

Zusammenfassung

Onkolytische Viren sind replikationsfähige „lebende“ Viren. Sie infizieren hochselektiv Tumorzellen, vermehren sich in diesen und zerstören sie dabei. Aufgrund der enormen Fortschritte auf dem Gebiet der Gen- und Biotechnologie kommt die Virotherapie zunehmend in klinischen Studien zum Einsatz und erweist sich als äußerst sicher, nebenwirkungsarm und effektiv. Insbesondere findet sich bei der Behandlung von Ovarialkarzinompatientinnen ein wichtiger Forschungsschwerpunkt. Zum einen liegt dies an der schlechten Prognose der Erkrankung und der hieraus resultierenden Notwendigkeit neuer Therapiemodalitäten. Zum anderen breitet sich das Ovarialkarzinom typischerweise lokoregionär aus, woraus sich die spezielle Möglichkeit der intraperitonealen Applikation onkolytischer Viren ergibt. Die vorliegende Arbeit fasst die vielversprechenden Ergebnisse klinischer Studien zur Behandlung von Ovarialkarzinompatientinnen mit onkolytischen Viren zuammen.

Abstract

Oncolytic viruses are replication competent “live” viruses. They infect tumor cells, replicate highly selective inside and thereby destroy them. Because of the enormous advances in the field of genetic engineering and biotechnology during the last decade, virotherapy is increasingly used within clinical trials and proved to be safe and effective. In particular, treatment of ovarian cancer patients is one main focus of research. On the one hand, this is due to the poor prognosis of this dismal entity, resulting in the urgent need for novel therapeutics. On the other hand, as ovarian cancer typically spreads within the peritoneal cavity, intraperitoneal administration of oncolytic viruses is feasible. This paper provides an overview of promising results from clinical trials to treat ovarian cancer patients with oncolytic viruses.

 
  • References

  • 1 Husmann G. Robert Koch-Inst Hrsg. Krebs in Deutschland: 2005/2006; Häufigkeiten und Trends; Eine gemeinsame Veröffentlichung des Robert Koch-Instituts und der Gesellschaft der Epidemiologischen Krebsregister in Deutschland e.V. Berlin: Robert Koch-Institut; 2010
  • 2 NIH consensus conference. Ovarian cancer. Screening, treatment, and follow-up. NIH Consensus Development Panel on Ovarian Cancer. JAMA 1995; 273: 491-497
  • 3 Heitz F, Harter P, Zelazny J et al. Antiangiogenic approaches in ovarian cancer therapy. Geburtsh Frauenheilk 2010; 70: 791-797
  • 4 Hartkopf AD, Fehm T, Wallwiener D et al. Oncolytic virotherapy of gynecologic malignancies. Gynecol Oncol 2011; 120: 302-310
  • 5 Bitzer M, Lauer UM. [Oncolytic viruses for genetic therapy of gastrointestinal tumors]. Z Gastroenterol 2003; 41: 667-674
  • 6 Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther 2007; 15: 651-659
  • 7 Bluming AZ, Ziegler JL. Regression of Burkittʼs lymphoma in association with measles infection. Lancet 1971; 2: 105-106
  • 8 Zygiert Z. Hodgkinʼs disease: remissions after measles. Lancet 1971; 1: 593
  • 9 Huebner RJ, Rowe WP, Schatten WE et al. Studies on the use of viruses in the treatment of carcinoma of the cervix. Cancer 1956; 9: 1211-1218
  • 10 Cassel WA, Garrett RE. Newcastle disease virus as an antineoplastic agent. Cancer 1965; 18: 863-868
  • 11 Liu TC, Galanis E, Kirn D. Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress. Nat Clin Pract Oncol 2007; 4: 101-117
  • 12 Kirn D, Martuza RL, Zwiebel J. Replication-selective virotherapy for cancer: Biological principles, risk management and future directions. Nat Med 2001; 7: 781-787
  • 13 Lorence RM, Rood PA, Kelley KW. Newcastle disease virus as an antineoplastic agent: induction of tumor necrosis factor-alpha and augmentation of its cytotoxicity. J Natl Cancer Inst 1988; 80: 1305-1312
  • 14 Peplinski GR, Tsung AK, Casey MJ et al. In vivo murine tumor gene delivery and expression by systemic recombinant vaccinia virus encoding interleukin-1beta. Cancer J Sci Am 1996; 2: 21-27
  • 15 Stojdl DF, Lichty B, Knowles S et al. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Nat Med 2000; 6: 821-825
  • 16 Geletneky K, Hartkopf AD, Krempien R et al. Improved killing of human high-grade glioma cells by combining ionizing radiation with oncolytic parvovirus H-1 infection. J Biomed Biotechnol 2010; 2010: 350748
  • 17 Zimmermann M, Armeanu S, Smirnow I et al. Human precision-cut liver tumor slices as a tumor patient-individual predictive test system for oncolytic measles vaccine viruses. Int J Oncol 2009; 34: 1247-1256
  • 18 Coffey MC, Strong JE, Forsyth PA et al. Reovirus therapy of tumors with activated Ras pathway. Science 1998; 282: 1332-1334
  • 19 Bauerschmitz G, Breidenbach M, Dall P et al. Mehrfach modifizierte onkolytische Adenoviren in der präklinischen Testung an Ovarialkarzinomzellen. Geburtsh Frauenheilk 2007; 67: 1102-1108
  • 20 Cutts FT, Markowitz LE. Successes and failures in measles control. J Infect Dis 1994; 170 (Suppl. 01) S32-S41
  • 21 Dorig RE, Marcil A, Chopra A et al. The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell 1993; 75: 295-305
  • 22 Anderson BD, Nakamura T, Russell SJ et al. High CD46 receptor density determines preferential killing of tumor cells by oncolytic measles virus. Cancer Res 2004; 64: 4919-4926
  • 23 Peng KW, Facteau S, Wegman T et al. Non-invasive in vivo monitoring of trackable viruses expressing soluble marker peptides. Nat Med 2002; 8: 527-531
  • 24 Galanis E, Hartmann LC, Cliby WA et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res 2010; 70: 875-882
  • 25 Milton GW, Brown MM. The limited role of attenuated smallpox virus in the management of advanced malignant melanoma. Aust N Z J Surg 1966; 35: 286-290
  • 26 Hunter-Craig I, Newton KA, Westbury G et al. Use of vaccinia virus in the treatment of metastatic malignant melanoma. Br Med J 1970; 2: 512-515
  • 27 Arakawa Jr. S, Hamami G, Umezu K et al. Clinical trial of attenuated vaccinia virus AS strain in the treatment of advanced adenocarcinoma. Report on two cases. J Cancer Res Clin Oncol 1987; 113: 95-98
  • 28 Gomella LG, Mastrangelo MJ, McCue PA et al. Phase i study of intravesical vaccinia virus as a vector for gene therapy of bladder cancer. J Urol 2001; 166: 1291-1295
  • 29 McCart JA, Ward JM, Lee J et al. Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Res 2001; 61: 8751-8757
  • 30 Hung CF, Tsai YC, He L et al. Vaccinia virus preferentially infects and controls human and murine ovarian tumors in mice. Gene Ther 2007; 14: 20-29
  • 31 Chalikonda S, Kivlen MH, OʼMalley ME et al. Oncolytic virotherapy for ovarian carcinomatosis using a replication-selective vaccinia virus armed with a yeast cytosine deaminase gene. Cancer Gene Ther 2008; 15: 115-125
  • 32 Hirasawa K, Nishikawa SG, Norman KL et al. Oncolytic reovirus against ovarian and colon cancer. Cancer Res 2002; 62: 1696-1701
  • 33 Lal R, Harris D, Postel-Vinay S et al. Reovirus: Rationale and clinical trial update. Curr Opin Mol Ther 2009; 11: 532-539
  • 34 Cohn DE, Nuovo G, Coffey MC et al. Phase I/II trial of reovirus serotype 3-Dearing strain in patients with recurrent ovarian cancer. J Clin Oncol 2010; 28: 15s (Abstr. TP253)
  • 35 Phelps MA, Cohn DE, OʼMalley DM et al. Reovirus replication in ovarian and peritoneal tumors after intravenous administration. Proceedings of the 101st Annual Meeting of the American Association for Cancer Research 2010; Abstract No. 2594
  • 36 Vollmer CM, Ribas A, Butterfield LH et al. p 53 selective and nonselective replication of an E1B-deleted adenovirus in hepatocellular carcinoma. Cancer Res 1999; 59: 4369-4374
  • 37 Bischoff JR, Kirn DH, Williams A et al. An adenovirus mutant that replicates selectively in p 53-deficient human tumor cells. Science 1996; 274: 373-376
  • 38 Yuan ZY, Zhang L, Li S et al. [Safety of an E1B deleted adenovirus administered intratumorally to patients with cancer]. Ai Zheng 2003; 22: 310-313
  • 39 Vasey PA, Shulman LN, Campos S et al. Phase I trial of intraperitoneal injection of the E1B-55-kd-gene-deleted adenovirus ONYX-015 (dl1520) given on days 1 through 5 every 3 weeks in patients with recurrent/refractory epithelial ovarian cancer. J Clin Oncol 2002; 20: 1562-1569
  • 40 Liu HL, Chen J. Oncolytic virus as an agent for the treatment of malignant ascites. Cancer Biother Radiopharm 2009; 24: 99-102
  • 41 Bauerschmitz GJ, Lam JT, Kanerva A et al. Treatment of ovarian cancer with a tropism modified oncolytic adenovirus. Cancer Res 2002; 62: 1266-1270
  • 42 Kimball KJ, Preuss MA, Barnes MN et al. A phase I study of a tropism-modified conditionally replicative adenovirus for recurrent malignant gynecologic diseases. Clin Cancer Res 2010; 16: 5277-5287
  • 43 Fisher K. Striking out at disseminated metastases: the systemic delivery of oncolytic viruses. Curr Opin Mol Ther 2006; 8: 301-313
  • 44 Hemminki A, Belousova N, Zinn KR et al. An adenovirus with enhanced infectivity mediates molecular chemotherapy of ovarian cancer cells and allows imaging of gene expression. Mol Ther 2001; 4: 223-231
  • 45 Lolkema MP, Arkenau HT, Harrington K et al. A phase I study of the combination of intravenous reovirus type 3 Dearing and gemcitabine in patients with advanced cancer. Clin Cancer Res 2011; 17: 581-588
  • 46 Fulci G, Breymann L, Gianni D et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad Sci USA 2006; 103: 12873-12878
  • 47 Mader EK, Maeyama Y, Lin Y et al. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res 2009; 15: 7246-7255
  • 48 Willmon C, Harrington K, Kottke T et al. Cell carriers for oncolytic viruses: Fed Ex for cancer therapy. Mol Ther 2009; 17: 1667-1676
  • 49 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: 4297-4309
  • 50 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: 1874-1884
  • 51 du Bois A, Schmalfeldt B, Meier W et al. Onkologie. Ovarialkarzinom – Ist die intraperitoneale Therapie wirklich neuer Standard?. Geburtsh Frauenheilk 2006; 66: 604-605
  • 52 Lage H. Mechanismen der Chemotherapieresistenz beim Ovarialkarzinom. Geburtsh Frauenheilk 2007; 67: 831-836
  • 53 Hartkopf AD, Fehm T, Wallwiener D et al. Oncolytic virotherapy of breast cancer. Gynecol Oncol 2011; 123: 164-171