Virology and Emerging Diseases - Sci Forschen

Full Text

Opinion Article
Cytomegalovirus and Cancer: A Long and Winding Road to Conclusive Evidence

  Jindrich Cinatl Jr1      Martin Michaelis2*  

1Institute of Medical Virology Goethe University, Paul Ehrlich-Str. 40, 60596 Frankfurt am Main, Germany
2Centre for Molecular Processing and School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom

*Corresponding author: Martin Michaelis, Centre for Molecular Processing and School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom, E-mail: M.Michaelis@kent.ac.uk

It has long been known that collecting convincing evidence relating a certain virus to the formation of one or more cancer type(s) is a tedious task. In 1911, Peyton Rous, being aware of similar previous experiments on the transmission of leukaemias between chicken by Vilhelm Ellerman and Olaf Bang [1], had shown that cell-free extracts from chicken sarcomas induced sarcomas in healthy chickens [2]. He concluded that a “minute parasitic organism” or a “chemical stimulant, elaborated by the neoplastic cells” must be responsible for sarcoma formation [2]. Later, the causative agent was identified to be a retrovirus and called the Rous sarcoma virus (RSV) [3]. However, it took 55 years and the discovery of the Epstein-Barr virus, the first human tumour virus [3,4], before Peyton Rouse eventually received the Nobel Prize for his work in 1966.

Since 1972, human cytomegalovirus (HCMV) has been associated with cancer in various ways [5-7]. In contrast to the eight viruses that are known to contribute to carcinogenesis in humans (Epstein-Barr virus, hepatitis B virus, human T-lymphotropic virus-I, high risk human papilloma viruses, hepatitis C virus, Kaposi’s sarcoma herpesvirus, Merkel cell polyomavirus) [8], HCMV is not regarded as a tumour virus because evidence that it can transform human cells is lacking [6,7].

To describe a possible interaction of cytomegalovirus (and potentially other viruses) with cancer cells, the concept of oncomodulation was developed based on experimental findings [9,10] and a neuroblastoma patient with therapy-refractory disease who had developed HCMV disease and in whom antiviral therapy with ganciclovir not only suppressed HCMV replication but also resulted in resensitisation of the tumour to anti-cancer therapy [7].

Oncomodulation means that a virus enhances the malignancy of established cancer cells independently of its carcinogenic potential [6,7,9,10]. Oncomodulatory effects are also exerted by tumour viruses, but HCMV turned out to be an ideally suited model virus to study oncomodulation by viruses because it does not transform cells. A number of different groups determined oncomodulatory effects by HCMV in a wide range of experimental systems [6,7,11-14].

Interest into the clinical role of HCMV in cancer sharply increased from 2002 onwards, after the group of Charles Cobbs had detected HCMV at very high frequencies in cancer tissues from glioma patients [15], colorectal cancer patients [16], and prostate cancer patients [17] by so-called ‘highly sensitive’ detection methods [18,19]. A couple of groups confirmed these findings, and to date the presence of HCMV has been reported in very large fractions of investigated glioma, colorectal cancer, prostate cancer, breast cancer, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and hepatocellular cancer tissues [6,7,14,20-27]. With regard to the investigation of HCMV in cancer, glioma has been the most intensively studied cancer entity [6,7,14,20]. This includes studies that not only indicate the presence of HCMV DNA, RNA, and/ or proteins in cancer cells but also HCMV-induced signalling events and correlations suggesting that higher levels of HCMV infection of tumour tissues are associated with advanced disease stages and poorer outcomes [6,7,14,24,26,28,29]. Moreover, HCMV pp65-specific T-cells were reported to kill autologous primary glioblastoma cells, and dendritic cells pulsed with tumour RNA were described to cause an expansion of HCMV pp65-specific T-cells [30].

However, a significant number of other research groups failed to detect HCMV in cancers including glioma, neuroblastoma, childhood brain tumours, breast cancer, colorectal cancer, cervical cancer, gastric cancer, head and neck cancer, renal cancer, leukaemias, liver cancer, ovarian cancer, prostate cancer, and skin cancer [6,7,31-42). Most strikingly, no deep sequencing study has been able to detect HCMV in cancer so far [34,36,37,42]. Thus, there is an ongoing dispute whether HCMV is actually present in cancer tissues or not.

A concern with regard to the presence of HCMV in glioma is that there is no consistent relationship between the HCMV seroprevalence and the glioma incidence. Gliomas are not overrepresented among HCMV-seropositive individuals, and the prognosis does not appear to differ between HCMV-seropositive and –seronegative glioma patients [35,43,44]. One study had reported a significant proportion of patients (80%) with newly diagnosed glioblastoma to have detectable HCMV DNA in their peripheral blood, while sero-positive normal donors and other surgical patients did not exhibit detectable virus [45]. The authors interpreted this as either being a systemic reactivation of HCMV within glioblastoma patients or virus shedding from infected cancer cells [45]. However, others failed to detect enhanced HCMV DNA levels in glioblastoma patients [46,47].

Notably, the ‘highly sensitive methods’ detect HCMV in tumour tissues from HMCV-seropositive and –seronegative patients [48-50]. The authors of one study concluded that HCMV seropositivity is not indicative for the presence of HCMV in tumour tissues [48]. Indeed, one study correlated high anti-HCMV IgG with a low glioblastoma risk and low anti-HCMV IgG with a high glioblastoma risk [51]. However, this appears to be in conflict with findings showing that there is no difference in disease outcome between HCM-seropositive and HCMVseronegative glioblastoma patients [35,43,44]. It remains also unclear how HCMV should reach tumour tissues without inducing an immune response. Serology was criticised to be a ‘poor test’ for detecting HCMV in glioblastoma patients [50]. However, the currently applied serological methods are highly reliable in identifying the (HCMV-positive) immunocompromised patients that are at risk of HCMV disease [52].

Despite these controversies, clinical trials have started that aim to exploit HCMV as a therapeutic target for the treatment of glioblastoma patients. Initial trials on HCMV-directed immunotherapies for glioblastoma patients indicated these approaches to be safe but efficacy data is not available, yet [53,54]. Arandomised trial investigating the antiHCMV drug valganciclovirin glioblastoma patients did not demonstrate any benefit over placebo [55]. Subsequent re-analysis of parts of this study in combination with some additional cases, however, demonstrated a survival advantage for glioblastoma patients treated with valganciclovir [56,57]. However, this approach was strongly criticised as being flawed [58-61]. It is possible that a compound like ganciclovir, the active principle of the prodrug valganciclovir, exerts HCMV-independent anti-cancer effects. The anti-HCMV drug cidofovir was demonstrated to exert antiglioblastoma effects in non-HCMV infected cancer cells [62] and similar unpublished results have been reported for ganciclovir [63].

Taken together, the question whether HCMV may play a role in cancer has been around since the 1970s [6] the concept of oncomodulation was proposed for the first time almost 20 years ago [9,10], the presence of HCMV has been suggested in large fraction of tumours from various cancer types since 2002 [14,15]. We have been confronted with requests from desperate cancer patients who wanted to know whether they should be treated with antiviral drugs for almost two decades now, and we still do not have an answer to this question. This is very frustrating. Arguably, the situation may be even more complex. For example, although we initially assumed that oncomodulation by HCMV depended on the presence of HCMV (antigens) in the tumour tissue [6], our later experimental findings indicated that HCMV can irreversibly increase cancer cell malignancy [64]. Also, HCMV-induced γδ T cells were described to exert anti-cancer effects [65,66]. However, today we are still not even able to answer with certainty the relative easy question whether HCMV is present in patient tumour tissues. This should be a wake-up call.

References
  1. Ellerman V, Bang O (1908) Experimentelle Leukämie bei Hühnern. Zentralbl Bakteriol Parasitenkd Infections krHyg Abt Orig 46: 595-609.
  2. Rous P (1911) A sarcoma of the fowl transmissible by an agent separable from the tumor cells. J Exp Med 13: 397-411. [Ref.]
  3. Weiss RA, Vogt PK (2011) 100 years of Rous sarcoma virus. J Exp Med 208: 2351-2355. [Ref.]
  4. Epstein MA, Achong BG, Barr YM (1964) Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet 1: 702-703. [Ref.]
  5. Vestergaard BF, Hornsleth A, Pedersen SN (1972) Occurrence of herpes- and adenovirus antibodies in patients with carcinoma of the cervix uteri. Measurement of antibodies to herpesvirus hominis (types 1 and 2), cytomegalovirus, EB-virus, and adenovirus. Cancer 30: 68- 74. [Ref.]
  6. Michaelis M, Doerr HW, Cinatl J Jr (2009) The story of human cytomegalovirus and cancer: increasing evidence and open questions. Neoplasia 11: 1-9. [Ref.]
  7. Michaelis M, Baumgarten P, Mittelbronn M, Driever PH, Doerr HW, et al. (2011) Oncomodulation by human cytomegalovirus: novel clinical findings open new roads. Med Microbiol Immunol 200: 1-5. [Ref.]
  8. Moore PS, Chang Y (2010) Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer 10: 878-889. [Ref.]
  9. Cinatl J Jr, Cinatl J, Vogel JU, Rabenau H, Kornhuber B, et al. (1996) Modulatory effects of human cytomegalovirus infection on malignant properties of cancer cells. Intervirology 39: 259-269. [Ref.]
  10. Cinatl J Jr, Vogel JU, Cinatl J, Weber B, Rabenau H, et al. (1996) Long-term productive human cytomegalovirus infection of a human neuroblastoma cell line. Int J Cancer 65: 90-96.
  11. Bishop RK, Valle Oseguera CA, Spencer JV (2015) Human Cytomegalovirus interleukin-10 promotes proliferation and migration of MCF-7 breast cancer cells. Cancer Cell Microenviron 2: e678. [Ref.]
  12. wahori S, Hakki M, Chou S, Kalejta RF (2015) Molecular Determinants for the Inactivation of the Retinoblastoma Tumor Suppressor by the Viral Cyclin-dependent Kinase UL97. J Biol Chem 290: 19666-19880. [Ref.]
  13. Price RL, Chiocca EA (2015) Modeling cytomegalovirus infection in mouse tumor models. Front Oncol 5: 61. [Ref.]
  14. Söderberg-Nauclér C, Johnsen JI (2015) Cytomegalovirus in human brain tumors: Role in pathogenesis and potential treatment options. World J Exp Med 5: 1-10. [Ref.]
  15. Cobbs CS, Harkins L, Samanta M, Gillespie GY, Bharara S, et al. (2002) Human cytomegalovirus infection and expression in human malignant glioma. Cancer Res 62: 3347-3350. [Ref.]
  16. Harkins L, Volk AL, Samanta M, Mikolaenko I, Britt WJ, et al. (2002) Specific localisation of human cytomegalovirus nucleic acids and proteins in human colorectal cancer. Lancet 360: 1557-1563. [Ref.]
  17. Samanta M, Harkins L, Klemm K, Britt WJ, Cobbs CS (2003) High prevalence of human cytomegalovirus in prostatic intraepithelial neoplasia and prostatic carcinoma. J Urol 170: 998-1002. [Ref.]
  18. Cobbs CS (2014) Response to “Human cytomegalovirus infection in tumor cells of the nervous system is not detectable with standardized pathologico-virological diagnostics”. Neuro Oncol 16: 1435-1436. [Ref.]
  19. Cobbs CS, Matlaf L, Harkins LE (2014) Methods for the detection of cytomegalovirus in glioblastoma cells and tissues. Methods Mol Biol 1119: 165-196. [Ref.]
  20. Dziurzynski K, Chang SM, Heimberger AB, Kalejta RF, McGregor Dallas SR, et al. (2012) Consensus on the role of human cytomegalovirus in glioblastoma. Neuro Oncol 14: 246-255. [Ref.]
  21. Ranganathan P, Clark PA, Kuo JS, Salamat MS, Kalejta RF (2012) Significant association of multiple human cytomegalovirus genomic Loci with glioblastoma multiforme samples. J Virol 86: 854-864. [Ref.]
  22. Wolmer-Solberg N, Baryawno N, Rahbar A, Fuchs D, Odeberg J, et al. (2013) Frequent detection of human cytomegalovirus in neuroblastoma: a novel therapeutic target? Int J Cancer 133: 2351- 2361. [Ref.]
  23. Chen HP, Jiang JK, Chan CH, Teo WH, Yang CY, et al. (2015) Genetic polymorphisms of the human cytomegalovirus UL144 gene in colorectal cancer and its association with clinical outcome. J Gen Virol. [Ref.]
  24. Fornara O, Bartek J Jr, Rahbar A, Odeberg J, Khan Z, et al. (2015) Cytomegalovirus infection induces a stem cell phenotype in human primary glioblastoma cells: prognostic significance and biological impact. Cell Death Differ. [Ref.]
  25. Shamran HA, Kadhim HS, Hussain AR, Kareem A, Taub DD, et al. (2015) Detection of human cytomegalovirus in different histopathological types of glioma in Iraqi patients. BioMed Res Int 2015: 642652. [Ref.]
  26. Soroceanu L, Matlaf L, Khan S, Akhavan A, Singer E, et al. (2015) Cytomegalovirus Immediate-Early Proteins Promote Stemness Properties in Glioblastoma. Cancer Res 75: 3065-3076. [Ref.]
  27. Wakefield A, Pignata A, Ghazi A, Ashoori A, Hegde M, et al. (2015) Is CMV a target in pediatric glioblastoma? Expression of CMV proteins, pp65 and IE1-72 and CMV nucleic acids in a cohort of pediatric glioblastoma patients. J Neurooncol. [Ref.]
  28. Rahbar A, Stragliotto G, Orrego A, Peredo I, Taher C, et al. (2012) Low levels of Human Cytomegalovirus Infection in Glioblastoma multiforme associates with patient survival; -a case-control study. Herpesviridae 3: 3. [Ref.]
  29. Rahbar A, Orrego A, Peredo I, Dzabic M, Wolmer-Solberg N, et al. (2013) Human cytomegalovirus infection levels in glioblastoma multiforme are of prognostic value for survival. J Clin Virol 57: 36-42. [Ref.]
  30. Nair SK, De Leon G, Boczkowski D, Schmittling R, Xie W, et al. (2014) Recognition and killing of autologous, primary glioblastoma tumor cells by human cytomegalovirus pp65-specific cytotoxic T cells. Clin Cancer Res 20: 2684-2694. [Ref.]
  31. Knösel T, Schewe C, Dietel M, Petersen I (2004) Cytomegalovirus is not associated with progression and metastasis of colorectal cancer. Cancer Lett 211: 243-247. [Ref.]
  32. Sabatier J, Uro-Coste E, Pommepuy I, Labrousse F, Allart S, et al. (2005) Detection of human cytomegalovirus genome and gene products in central nervous system tumours. Br J Cancer 92: 747-750. [Ref.]
  33. Sehic D, Forslund O, Sandén E, Mengelbier LH, Karlsson J, et al. (2013) Absence of Epstein-Barr and cytomegalovirus infection in neuroblastoma cells by standard detection methodologies. Pediatr Blood Cancer 60: E91-E93. [Ref.]
  34. Tang KW, Alaei-Mahabadi B, Samuelsson T, Lindh M, Larsson E (2013) The landscape of viral expression and host gene fusion and adaptation in human cancer. Nat Commun 4: 2513. [Ref.]
  35. Baumgarten P, Michaelis M, Rothweiler F, Starzetz T, Rabenau HF, et al. (2014) Human cytomegalovirus infection in tumor cells of the nervous system is not detectable with standardized pathologicovirological diagnostics. Neuro Oncol 16: 1469-1477. [Ref.]
  36. Cimino PJ, Zhao G, Wang D, Sehn JK, Lewis JS Jr, et al. (2014) Detection of viral pathogens in high grade gliomas from unmapped next-generation sequencing data. Exp Mol Pathol 96: 310-315. [Ref.]
  37. Cosset É, Petty TJ, Dutoit V, Cordey S, Padioleau I, et al. (2014) Comprehensive metagenomic analysis of glioblastoma reveals absence of known virus despite antiviral-like type I interferon gene response. Int J Cancer 135: 1381-1389. [Ref.]
  38. Forslund O, Holmquist Mengelbier L, Gisselsson D (2014) Regarding human cytomegalovirus in neuroblastoma. Cancer Med 3: 1038-1040. [Ref.]
  39. Yamashita Y, Ito Y, Isomura H, Takemura N, Okamoto A, et al. (2014) Lack of presence of the human cytomegalovirus in human glioblastoma. Mod Pathol 27: 922-929. [Ref.]
  40. Richardson AK, Currie MJ, Robinson BA, Morrin H, Phung Y, et al. (2015) Cytomegalovirus and Epstein-Barr virus in breast cancer. PLoS One 10: e0118989. [Ref.]
  41. Sardi I, Lucchesi M, Becciani S, Facchini L, Guidi M, et al. (2015) Absence of human cytomegalovirus infection in childhood brain tumors. Am J Cancer Res 5: 2476-2483. [Ref.]
  42. Tang KW, Hellstrand K, Larsson E (2015) Absence of cytomegalovirus in high-coverage DNA sequencing of human glioblastoma multiforme. Int J Cancer 136: 977-981. [Ref.]
  43. Lehrer S, Green S, Ramanathan L, Rosenzweig K, Labombardi V (2012) No consistent relationship of glioblastoma incidence and cytomegalovirus seropositivity in whites, blacks, and Hispanics. Anticancer Res 32: 1113-1115. [Ref.]
  44. Priel E, Wohl A, Teperberg M, Nass D, Cohen ZR (2015) Human cytomegalovirus viral load in tumor and peripheral blood samples of patients with malignant gliomas. J Clin Neurosci 22: 326-330. [Ref.]
  45. Mitchell DA, Xie W, Schmittling R, Learn C, Friedman A, et al. (2008) Sensitive detection of human cytomegalovirus in tumors and peripheral blood of patients diagnosed with glioblastoma. Neuro Oncol 10: 10-18. [Ref.]
  46. Lehrer S, Labombardi V, Green S, Pessin-Minsley MS, Germano IM, et al. (2011) No circulating cytomegalovirus in five patients with glioblastoma multiforme. Anticancer Res 31: 959-960. [Ref.]
  47. Lehrer S, Green S, Rosenzweig KE, Rendo A (2015) No circulating human cytomegalovirus in 14 cases of glioblastoma. Neuro Oncol 17: 320. [Ref.]
  48. Bianchi E, Roncarati P, Hougrand O, Guérin-El Khourouj V, Boreux R, et al. (2015) Human cytomegalovirus and primary intracranial tumours: frequency of tumour infection and lack of correlation with systemic immune anti-viral responses. Neuropathol Appl Neurobiol 41: e29-e40. [Ref.]
  49. Michaelis M, Mittelbronn M, Cinatl J Jr (2015) Towards an unbiased, collaborative effort to reach evidence about the presence of human cytomegalovirus in glioblastoma (and other tumors). Neuro Oncol 17: 1039. [Ref.]
  50. Rahbar A, Peredo I, Solberg NW, Taher C, Dzabic M, et al. (2015) Discordant humoral and cellular immune responses to Cytomegalovirus (CMV) in glioblastoma patients whose tumors are positive for CMV. Oncoimmunology 4: e982391. [Ref.]
  51. Amirian ES, Marquez-Do D, Bondy ML, Scheurer ME (2013) Antihuman-cytomegalovirus immunoglobulin G levels in glioma risk and prognosis. Cancer Med 2: 57-62. [Ref.]
  52. Einsele H, Mielke S, Grigoleit GU (2014) Diagnosis and treatment of cytomegalovirus 2013. Curr Opin Hematol 21: 470-475. [Ref.]
  53. Schuessler A, Smith C, Beagley L, Boyle GM, Rehan S, et al. (2014) Autologous T-cell therapy for cytomegalovirus as a consolidative treatment for recurrent glioblastoma. Cancer Res 74: 3466-3476. [Ref.]
  54. Mitchell DA, Batich KA, Gunn MD, Huang MN, Sanchez-Perez L, et al. (2015) Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 519: 366-369. [Ref.]
  55. Stragliotto G, Rahbar A, Solberg NW, Lilja A, Taher C, et al. (2013) Effects of valganciclovir as an add-on therapy in patients with cytomegalovirus-positive glioblastoma: a randomized, double-blind, hypothesis-generating study. Int J Cancer 133: 1204-1213. [Ref.]
  56. Söderberg-Nauclér C, Rahbar A, Stragliotto G (2013) Survival in patients with glioblastoma receiving valganciclovir. N Engl J Med 369: 985-986. [Ref.]
  57. Söderberg-Naucler C, Peredo I, Rahbar A, Hansson F, Nordlund A, et al. (2014) Use of Cox regression with treatment status as a timedependent covariate to re-analyze survival benefit excludes immortal time bias effect in patients with glioblastoma who received prolonged adjuvant treatment with valganciclovir. Int J Cancer 135: 248-249. [Ref.]
  58. Liu CJ, Hu YW (2014) Immortal time bias in retrospective analysis: is there a survival benefit in patients with glioblastoma who received prolonged treatment of adjuvant valganciclovir? Int J Cancer 135: 250-251. [Ref.]
  59. Weller M, Soffietti R, Brada M (2014) The legend of cytomegalovirus and glioblastoma lives on. Neuro Oncol 16: 166. [Ref.]
  60. Wick W, Platten M (2014) CMV infection and glioma, a highly controversial concept struggling in the clinical arena. Neuro Oncol 16: 332-333. [Ref.]
  61. Wick W, Wick A, Platten M (2014) Challenging cytomegalovirus data in glioblastoma. Neuro Oncol 16: 165. [Ref.]
  62. Hadaczek P, Ozawa T, Soroceanu L, Yoshida Y, Matlaf L, et al. (2013) Cidofovir: a novel antitumor agent for glioblastoma. Clin Cancer Res 19: 6473-6483. [Ref.]
  63. Cobbs CS (2014) Does valganciclovir have a role in glioblastoma therapy? Neuro Oncol 16: 330-331. [Ref.]
  64. Michaelis M, Barth S, Breitling R, Bruch J, Steinberger D, et al. (2012) Selection of a highly invasive neuroblastoma cell population through long-term human cytomegalovirus infection. Oncogenesis 1: e10. [Ref.]
  65. Devaud C, Bilhere E, Loizon S, Pitard V, Behr C, et al. (2009) Antitumor activity of gamma delta T cells reactive against cytomegalovirusinfected cells in a mouse xenograft tumor model. Cancer Res 69: 3971-3978. [Ref.]
  66. Couzi L, Pitard V, Moreau JF, Merville P, Déchanet-Merville J (2015) Direct and Indirect Effects of Cytomegalovirus-Induced γδ T Cells after Kidney Transplantation. Front Immunol 6: 3. [Ref.]

Download Provisional PDF Here

 

Article Information

Article Type: Opinion Article

Citation: Cinatl J Jr., Michaelis M (2015) Cytomegalovirus and Cancer: A Long and Winding Road to Conclusive Evidence. J Emerg Dis Virol 1 (2): doi http://dx.doi.org/10.16966/2473-1846.106

Copyright: © 2015 Cinatl J Jr., et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Publication history: 

  • Received date: 14 October 2015

  • Accepted date: 20 October 2015

  • Published date: 22 october 2015
  •