National Cancer Institute
U.S. National Institutes of Health | www.cancer.gov

NCI Home
Cancer Topics
Clinical Trials
Cancer Statistics
Research & Funding
News
About NCI
Newcastle Disease Virus (PDQ®)
Patient Version   Health Professional Version   Last Modified: 04/24/2008



Purpose of This PDQ Summary






Overview






General Information






History






Laboratory/Animal/Preclinical Studies






Human/Clinical Studies






Adverse Effects






Overall Level of Evidence for Newcastle Disease Virus






Changes to This Summary (04/24/2008)






More Information



Page Options
Print This Page
Print Entire Document
View Entire Document
E-Mail This Document
Quick Links
Director's Corner

Dictionary of Cancer Terms

NCI Drug Dictionary

Funding Opportunities

NCI Publications

Advisory Boards and Groups

Science Serving People

Español
Quit Smoking Today
NCI Highlights
Report to Nation Finds Declines in Cancer Incidence, Death Rates

High Dose Chemotherapy Prolongs Survival for Leukemia

Prostate Cancer Study Shows No Benefit for Selenium, Vitamin E

The Nation's Investment in Cancer Research FY 2009

Past Highlights
Laboratory/Animal/Preclinical Studies

Effects of Newcastle Disease Virus on Human Cancer Cells
NDV and Cancer Immunotherapy



Effects of Newcastle Disease Virus on Human Cancer Cells

The ability of Newcastle disease virus (NDV) to replicate efficiently in human cancer cells has been demonstrated in both laboratory studies and animal studies.[1-12] Reviewed in [13,14] Several of these studies have provided much of the evidence that lytic strains of NDV are also oncolytic.[3-6,8-10,12] Reviewed in [13]

Strain 73-T, which is lytic, has been shown to kill the following types of human cancer cells in vitro : fibrosarcoma, osteosarcoma, neuroblastoma, bladder carcinoma, cervical carcinoma, melanoma, Wilms tumor, and myeloid leukemia;[3,6,8,9] however, this strain did not kill human B-cell lymphoma (i.e., Burkitt lymphoma) cells in vitro.[8] In addition, strain 73-T did not kill normal, proliferating human white blood cells or normal human skin fibroblasts in vitro,[3,6,8] but it killed normal human lung fibroblasts in vitro at the same rate that it killed cancer cells.[8]

Lytic strain Roakin has been reported to kill human B-cell lymphoma and human T-cell lymphoma (i.e., Hodgkin lymphoma) cells in vitro four to five times faster than it kills normal, resting human white blood cells.[4,5] This strain, however, has also been reported to kill normal, proliferating human white blood cells in vitro, though at a lower rate than it kills cancer cells.[4]

Lytic strain Italien (or Italian) has been shown to kill human squamous cell lung carcinoma, melanoma, breast carcinoma, and larynx carcinoma, but not cervical carcinoma, cells in vitro.[12] The replication efficiency of this strain in normal human cells was not tested.

Overall, these results show that there are some types of human cancer cells in which individual lytic strains of NDV do not replicate very well and that there are some types of normal human cells in which they replicate very efficiently. Nonetheless, these data and the absence of serious illness in individuals infected with NDV Reviewed in [1-3,10,13,15-21] are consistent with the view that NDV replicates much more efficiently in human cancer cells than it does in most types of normal human cells.

NDV strain Ulster, which is nonlytic, has also been shown to replicate efficiently in human cancer cells in vitro, including cells of the following types of human tumors: colorectal carcinoma, gastric carcinoma, pancreatic carcinoma, bladder carcinoma, breast carcinoma, ovarian carcinoma, renal cell carcinoma, lung carcinoma, larynx carcinoma, cervical carcinoma, glioblastoma, melanoma, B-cell lymphoma, and T-cell lymphoma.[7] This strain does not replicate very efficiently in resting or proliferating normal human white blood cells in vitro.[7] Other experiments have shown that NDV Ulster can kill infected cells,[22] Reviewed in [14] and it can replicate in human cancer cells whether they are actively proliferating or not.[7] Reviewed in [20]

The ability of lytic strains of NDV to kill human cancer cells in vivo has also been examined. In xenograft studies, human cancer cells were injected either subcutaneously or intradermally into athymic, nude mice (i.e., mice that do not reject tumor cells from other animals because they have a defective immune system), and tumors were allowed to form. NDV was injected directly into the tumors, and tumor growth and animal survival were monitored.

Intratumoral injection of strain 73-T was associated with complete tumor regression in 75% to 100% of mice bearing human fibrosarcoma, neuroblastoma, or cervical carcinoma tumors.[1-3,10] Intratumoral injection of 73-T was also associated with more than 80% tumor regression in 66% of mice bearing human synovial sarcoma tumors.[2] In addition, intratumoral injection of 73-T was associated with 68% to 96% inhibition of tumor growth in mice bearing human epidermoid, colon, lung, breast, or prostate carcinoma tumors.[10]

Intratumoral injection of strain Italien was associated with complete tumor regression in 100% of mice bearing human melanoma tumors. The growth of metastatic tumors in these animals, however, was not affected, suggesting that the virus was unable to disseminate widely throughout the body.[11] Reviewed in [14,20]

Replication of strain 73-T in the above-mentioned neuroblastoma xenografts was demonstrated by showing an increase in the amount of virus that could be recovered from tumor samples over time.[1] When this strain was injected into the thigh muscle of athymic, nude mice, the amount of virus that could be recovered decreased with time,[1] a finding consistent with the proposal that NDV replicates much more efficiently in cancer cells than in most normal cells.

In one study, mice bearing human neuroblastoma xenografts were given single intraperitoneal injections of strain 73-T, and 9 (75%) of 12 treated mice exhibited complete, durable tumor regressions.[10]

It is important to note that athymic, nude mice still make small numbers of T cells, and they produce interferons, natural killer cells, and macrophages. Reviewed in [11,23,24] The possibility that these residual components of the immune system, which may be activated by the presence of NDV, contributed to the antitumor effects observed in the xenograft studies cannot be ruled out.

NDV and Cancer Immunotherapy

Other laboratory and animal studies have shown that NDV and NDV-infected cancer cells can stimulate a variety of immune system responses that are essential to the successful immunotherapy of cancer.[6,8,22,25-37] Reviewed in [11,20,38-42] A few of these studies used human cells,[6,8,26,27,35] Reviewed in [20,39,42] but most used animal cells and animal tumor models.[6,8,22,25,27-34,36] Reviewed in [11,20,38-41]

Data from a 2004 pilot clinical trial of an NDV-modified autologous tumor vaccine in 20 patients with stage III or IV head and neck squamous cell carcinomas suggest that the vaccine strategy can stimulate human antitumor immune responses in a manner similar to those found in animal models and may significantly prolong 5-year survival rates in this patient population. The study demonstrated the feasibility and safety of the vaccine regimen: no major side effects were observed in any of the patients.[43]

Two in vitro studies have shown that infection of human immune system cells with NDV causes the cells to produce and release the cytokines interferon-alpha and tumor necrosis factor (TNF)-alpha.[6,8] In one of these studies,[6] it was shown further that infection of human cancer cells with NDV makes the cells more sensitive to the cytotoxic effects of TNF-alpha.

Additional in vitro studies have shown that NDV-infected human cancer cells are better at activating human cytotoxic T cells, helper T cells, and natural killer cells than uninfected cancer cells.[8,26,27] The NDV protein hemagglutinin-neuraminidase, which is present in the plasma membrane of virus-infected cells, appears to play a role in the enhancement of T cell activation. There is evidence that this protein makes infected cells more adhesive, thereby promoting the interaction between virus-infected cells and immune system cells.[27] Reviewed in [20]

Other laboratory studies have shown that the interaction between NDV-infected cancer cells and T cells can be improved if monoclonal antibodies that bind the hemagglutinin-neuraminidase protein on the cancer cells and either the CD3 protein or the CD28 protein on T cells (i.e., bispecific monoclonal antibodies) are also used.[26,35] Reviewed in [20,39,42] It has been reported that this improved interaction leads to better T cell activation.[26,35] Reviewed in [20,39,42] T cells exposed to NDV-infected human colon cancer cells and bispecific monoclonal antibodies showed not only an increased ability to kill the virus-infected cells but also an ability to inhibit the proliferation of uninfected colon cancer cells.[26,35] Reviewed in [20] On the basis of these and other in vitro findings, it has been proposed that vaccines consisting of NDV-infected cancer cells and bispecific monoclonal antibodies be tested in humans.[20,26,35,39,42]

As noted above, animal cells and animal tumor models have also been used to explore the immunotherapy potential of NDV. ESb, a mouse model of metastatic T-cell lymphoma has been employed in most of this work;[22,25,28,32-34,36,37] Reviewed in [11,20,38-42] however, additional experiments have utilized one or more of the following tumor models: mouse B16 melanoma,[30] mouse Lewis lung carcinoma,[29,32] mouse P815 mastocytoma,[32] mouse Ca 761-P93 mammary carcinoma,[32] and guinea pig L10 hepatocellular carcinoma.[31]

In one study,[32] it was shown that anticancer activity could be induced in mouse macrophages both in vitro and in vivo by infection with NDV strain Ulster. Similar activation of mouse macrophages in vitro was observed after infection with the NDV lytic strain Lasota. In this study, the activated macrophages showed cytotoxic activity toward ESb, P815 mastocytoma, and Ca 761-P93 mammary carcinoma cells in vitro. Other experiments demonstrated that much of the observed anticancer activity could be attributed to the production and release of TNF-alpha by the infected macrophages. In addition, the infected, activated macrophages showed anticancer activity in vivo when they were injected into mice bearing Ca 761-P93 mammary carcinoma or Lewis lung carcinoma tumors.[32]

In another study, Reviewed in [11] intratumoral injection of NDV strain Ulster into growing ESb tumors in immunocompetent mice led to a cessation of tumor growth and an absence of metastases in 42% of treated animals. In the remaining mice, tumor growth and metastatic spread continued at the same rate as in control animals. Reviewed in [11] Additional results from this study indicated that the anticancer effect in the responding animals was due primarily to the activation of T cells directed against a tumor-specific antigen on ESb cells rather than a virus antigen. Reviewed in [11]

Other studies with NDV Ulster and the ESb tumor model support the idea that virus proteins inserted in the plasma membrane of NDV-infected cancer cells may help the immune system recognize tumor-specific antigens better, potentially leading to an increased ability to kill uninfected cancer cells and virus-infected cells.[22,25,28,33,34,36] Reviewed in [11,20,38,40,41] At least four studies [22,25,34,36] Reviewed in [40,41] have shown that T cells isolated from mice that have growing ESb tumors can be activated in vitro by co-culture with NDV-infected ESb cells and that the resulting activated T cells possess an enhanced ability to kill uninfected ESb cells in vitro. In addition, two in vivo studies [28] Reviewed in [11] have shown that mice injected with NDV-infected, irradiated ESb cells are 30 to 250 times more resistant to later injection with proliferating ESb cells than mice that are initially injected with uninfected, irradiated ESb cells. Furthermore, at least two in vivo studies have demonstrated that vaccination of mice with NDV-infected, irradiated ESb cells after surgery to remove a growing ESb primary tumor can prevent the growth of metastatic tumors in approximately 50% of treated animals.[28,33] Reviewed in [11,38,40,41] When the surviving mice were subsequently injected with proliferating ESb cells, they all remained free of cancer, indicating that the NDV/tumor cell vaccine had conferred anticancer immunity.[28,33] Reviewed in [11,40,41] Similar results were obtained from in vivo studies that employed the mouse B16 melanoma model,[30] the mouse Lewis lung carcinoma model,[29] or the guinea pig L10 hepatocellular carcinoma model.[31]

One factor that may influence the effectiveness of NDV/tumor cell vaccines is overall tumor burden. Results obtained with the B16 mouse melanoma model suggest that these vaccines are less effective in individuals with advanced metastatic disease.[30]

References

  1. Lorence RM, Reichard KW, Katubig BB, et al.: Complete regression of human neuroblastoma xenografts in athymic mice after local Newcastle disease virus therapy. J Natl Cancer Inst 86 (16): 1228-33, 1994.  [PUBMED Abstract]

  2. Lorence RM, Katubig BB, Reichard KW, et al.: Complete regression of human fibrosarcoma xenografts after local Newcastle disease virus therapy. Cancer Res 54 (23): 6017-21, 1994.  [PUBMED Abstract]

  3. Reichard KW, Lorence RM, Cascino CJ, et al.: Newcastle disease virus selectively kills human tumor cells. J Surg Res 52 (5): 448-53, 1992.  [PUBMED Abstract]

  4. Bar-Eli N, Giloh H, Schlesinger M, et al.: Preferential cytotoxic effect of Newcastle disease virus on lymphoma cells. J Cancer Res Clin Oncol 122 (7): 409-15, 1996.  [PUBMED Abstract]

  5. Tzadok-David Y, Metzkin-Eizenberg M, Zakay-Rones Z: The effect of a mesogenic and a lentogenic Newcastle disease virus strain on Burkitt lymphoma Daudi cells. J Cancer Res Clin Oncol 121 (3): 169-74, 1995.  [PUBMED Abstract]

  6. 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 80 (16): 1305-12, 1988.  [PUBMED Abstract]

  7. Schirrmacher V, Haas C, Bonifer R, et al.: Human tumor cell modification by virus infection: an efficient and safe way to produce cancer vaccine with pleiotropic immune stimulatory properties when using Newcastle disease virus. Gene Ther 6 (1): 63-73, 1999.  [PUBMED Abstract]

  8. Zorn U, Dallmann I, Grosse J, et al.: Induction of cytokines and cytotoxicity against tumor cells by Newcastle disease virus. Cancer Biother 9 (3): 225-35, 1994 Fall.  [PUBMED Abstract]

  9. Cassel WA, Garrett RE: Newcastle disease virus as an antineoplastic agent. Cancer 18: 863-8, 1965. 

  10. Phuangsab A, Lorence RM, Reichard KW, et al.: Newcastle disease virus therapy of human tumor xenografts: antitumor effects of local or systemic administration. Cancer Lett 172 (1): 27-36, 2001.  [PUBMED Abstract]

  11. Schirrmacher V, Ahlert T, Heicappell R, et al.: Successful application of non-oncogenic viruses for antimetastatic cancer immunotherapy. Cancer Rev 5: 19-49, 1986. 

  12. Ahlert T, Schirrmacher V: Isolation of a human melanoma adapted Newcastle disease virus mutant with highly selective replication patterns. Cancer Res 50 (18): 5962-8, 1990.  [PUBMED Abstract]

  13. Kirn DH, McCormick F: Replicating viruses as selective cancer therapeutics. Mol Med Today 2 (12): 519-27, 1996.  [PUBMED Abstract]

  14. Schirrmacher V, Griesbach A, Ahlert T: Antitumor effects of Newcastle Disease Virus in vivo: local versus systemic effects. Int J Oncol 18 (5): 945-52, 2001.  [PUBMED Abstract]

  15. Csatary LK, Moss RW, Beuth J, et al.: Beneficial treatment of patients with advanced cancer using a Newcastle disease virus vaccine (MTH-68/H). Anticancer Res 19 (1B): 635-8, 1999 Jan-Feb.  [PUBMED Abstract]

  16. Emergency Preparedness Information eXchange.: Foreign Animal Diseases: Newcastle Disease. Burnaby, B.C., Canada: Telematics Research Lab, Simon Fraser University, 2002. Available online. Last accessed May 2, 2006. 

  17. Csatary LK, Eckhardt S, Bukosza I, et al.: Attenuated veterinary virus vaccine for the treatment of cancer. Cancer Detect Prev 17 (6): 619-27, 1993.  [PUBMED Abstract]

  18. Kenney S, Pagano JS: Viruses as oncolytic agents: a new age for "therapeutic" viruses? J Natl Cancer Inst 86 (16): 1185-6, 1994.  [PUBMED Abstract]

  19. Batliwalla FM, Bateman BA, Serrano D, et al.: A 15-year follow-up of AJCC stage III malignant melanoma patients treated postsurgically with Newcastle disease virus (NDV) oncolysate and determination of alterations in the CD8 T cell repertoire. Mol Med 4 (12): 783-94, 1998.  [PUBMED Abstract]

  20. Schirrmacher V, Ahlert T, Pröbstle T, et al.: Immunization with virus-modified tumor cells. Semin Oncol 25 (6): 677-96, 1998.  [PUBMED Abstract]

  21. Moss RW: Alternative pharmacological and biological treatments for cancer: ten promising approaches. J Naturopathic Med 6 (1): 23-32, 1996. 

  22. Schirrmacher V, Jurianz K, Roth C, et al.: Tumor stimulator cell modification by infection with Newcastle Disease Virus: analysis of effects and mechanism in MLTC-CML cultures. Int J Oncol 14 (2): 205-15, 1999.  [PUBMED Abstract]

  23. Kadish AS, Doyle AT, Steinhauer EH, et al.: Natural cytotoxicity and interferon production in human cancer: deficient natural killer activity and normal interferon production in patients with advanced disease. J Immunol 127 (5): 1817-22, 1981.  [PUBMED Abstract]

  24. Budzynski W, Radzikowski C: Cytotoxic cells in immunodeficient athymic mice. Immunopharmacol Immunotoxicol 16 (3): 319-46, 1994.  [PUBMED Abstract]

  25. Schirrmacher V, Haas C, Bonifer R, et al.: Virus potentiation of tumor vaccine T-cell stimulatory capacity requires cell surface binding but not infection. Clin Cancer Res 3 (7): 1135-48, 1997.  [PUBMED Abstract]

  26. Haas C, Herold-Mende C, Gerhards R, et al.: An effective strategy of human tumor vaccine modification by coupling bispecific costimulatory molecules. Cancer Gene Ther 6 (3): 254-62, 1999 May-Jun.  [PUBMED Abstract]

  27. Haas C, Ertel C, Gerhards R, et al.: Introduction of adhesive and costimulatory immune functions into tumor cells by infection with Newcastle Disease Virus. Int J Oncol 13 (6): 1105-15, 1998.  [PUBMED Abstract]

  28. Heicappell R, Schirrmacher V, von Hoegen P, et al.: Prevention of metastatic spread by postoperative immunotherapy with virally modified autologous tumor cells. I. Parameters for optimal therapeutic effects. Int J Cancer 37 (4): 569-77, 1986.  [PUBMED Abstract]

  29. Shoham J, Hirsch R, Zakay-Rones Z, et al.: Augmentation of tumor cell immunogenicity by viruses--an approach to specific immunotherapy of cancer. Nat Immun Cell Growth Regul 9 (3): 165-72, 1990.  [PUBMED Abstract]

  30. Plaksin D, Porgador A, Vadai E, et al.: Effective anti-metastatic melanoma vaccination with tumor cells transfected with MHC genes and/or infected with Newcastle disease virus (NDV). Int J Cancer 59 (6): 796-801, 1994.  [PUBMED Abstract]

  31. Bier H, Armonat G, Bier J, et al.: Postoperative active-specific immunotherapy of lymph node micrometastasis in a guinea pig tumor model. ORL J Otorhinolaryngol Relat Spec 51 (4): 197-205, 1989.  [PUBMED Abstract]

  32. Schirrmacher V, Bai L, Umansky V, et al.: Newcastle disease virus activates macrophages for anti-tumor activity. Int J Oncol 16 (2): 363-73, 2000.  [PUBMED Abstract]

  33. Schirrmacher V, Heicappell R: Prevention of metastatic spread by postoperative immunotherapy with virally modified autologous tumor cells. II. Establishment of specific systemic anti-tumor immunity. Clin Exp Metastasis 5 (2): 147-56, 1987 Apr-Jun.  [PUBMED Abstract]

  34. von Hoegen P, Zawatzky R, Schirrmacher V: Modification of tumor cells by a low dose of Newcastle disease virus. III. Potentiation of tumor-specific cytolytic T cell activity via induction of interferon-alpha/beta. Cell Immunol 126 (1): 80-90, 1990.  [PUBMED Abstract]

  35. Haas C, Strauss G, Moldenhauer G, et al.: Bispecific antibodies increase T-cell stimulatory capacity in vitro of human autologous virus-modified tumor vaccine. Clin Cancer Res 4 (3): 721-30, 1998.  [PUBMED Abstract]

  36. Von Hoegen P, Weber E, Schirrmacher V: Modification of tumor cells by a low dose of Newcastle disease virus. Augmentation of the tumor-specific T cell response in the absence of an anti-viral response. Eur J Immunol 18 (8): 1159-66, 1988.  [PUBMED Abstract]

  37. Schirrmacher V, Schild HJ, Gückel B, et al.: Tumour-specific CTL response requiring interactions of four different cell types and recognition of MHC class I and class II restricted tumour antigens. Immunol Cell Biol 71 ( Pt 4): 311-26, 1993.  [PUBMED Abstract]

  38. Schirrmacher V: Active specific immunotherapy: a new modality of cancer treatment involving the patient's own immune system. Onkologie 16: 290-6, 1993. 

  39. Haas C, Schirrmacher V: Immunogenicity increase of autologous tumor cell vaccines by virus infection and attachment of bispecific antibodies. Cancer Immunol Immunother 43 (3): 190-4, 1996.  [PUBMED Abstract]

  40. Schirrmacher V, von Hoegen P, Heicappell R: Virus modified tumor cell vaccines for active specific immunotherapy of micrometastases: expansion and activation of tumor-specific T cells. Prog Clin Biol Res 288: 391-9, 1989.  [PUBMED Abstract]

  41. Schirrmacher V, von Hoegen P, Heicappell R: Postoperative activation of tumor specific T cells by immunization with virus-modified tumor cells and effects on metastasis. Adv Exp Med Biol 233: 91-6, 1988.  [PUBMED Abstract]

  42. Schirrmacher V, Haas C: Modification of cancer vaccines by virus infection and attachment of bispecific antibodies. In: Walden P, Trefzer U, Sterry W, et al., eds.: Gene Therapy of Cancer. New York, NY: Plenum Press, 1998, pp 251-7. 

  43. Karcher J, Dyckhoff G, Beckhove P, et al.: Antitumor vaccination in patients with head and neck squamous cell carcinomas with autologous virus-modified tumor cells. Cancer Res 64 (21): 8057-61, 2004.  [PUBMED Abstract]

Back to Top

< Previous Section  |  Next Section >


A Service of the National Cancer Institute
Department of Health and Human Services National Institutes of Health USA.gov