Skip to main content
Advertisement
  • Loading metrics

The antiviral immune forces awaken in the cancer wars

Introduction

Viruses are the most abundant microorganisms on earth, and it is estimated that there are at least 10 times more viruses than bacteria in the human microbiome [1]. Unlike bacteria, many viruses can permanently settle in a host cell, are minimally influenced by environmental changes, and can even be inherited when infecting germline cells. Many viruses have coevolved with host species for millions of years and have developed mechanisms to evade immune recognition and maintain an equilibrium state with the host immune system. Recent studies have shown that breaking this immune equilibrium can activate the host antiviral immune responses [26].

Current cancer immunotherapies targeting immune checkpoint molecules have limited efficacy in treatment of noninflamed tumors (so-called “cold” tumors) that show few infiltrating T cells. The absence of T-cell infiltration is largely caused by the lack of tumor antigens, antigen presentation, and/or abundance of immunosuppressive cells in the tumor microenvironment (TME) [7]. To convert immune “cold” tumors to inflamed “hot” tumors, a variety of different strategies are currently under investigation, including bispecific antibodies, chimeric antigen receptor (CAR) T cells, oncolytic viruses, and cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) agonists [8, 9]. The high mutation loads in tumors often result in expression of mutation-associated neoantigens, which are recognized as foreign antigens to the host T cells. These neoantigens have been found to be associated with improved responses to immune checkpoint inhibitors (ICIs) [10]. Interestingly, when suppressed viral gene expression in a tumor is reactivated from the virome, they are similarly recognized as tumor neoantigens by host immune systems [6, 11]. These findings suggest that activating immune responses against viruses in the human virome might be an effective tool for cancer immunotherapies.

Which viruses are included in the human virome for potential tumor neoantigens?

The human virome consists of a multitude of viruses, including eukaryotic viruses that infect human cells, viruses that infect prokaryotes (including bacteria and archaea), and viral genetic elements that reside within the host genome [12]. Although a significant portion of the human virome consists of bacteriophages, our current understanding of the virome mainly centers around eukaryotic viruses [13].

Human endogenous retroviruses (HERVs) are the most abundant viral elements in the human virome, making up over 8% of the human genome [14]. Previous studies have shown potential associations between increased HERV protein expression and human diseases [15, 16]. All HERVs exist in the human virome as permanently integrated elements in the host genome, which can be inherited by future generations through germline cells.

Similar to HERVs, human herpesvirus 6 (HHV-6) is integrated into the host germline chromosome and inherited in about 1% of human populations [17]. Several other types of herpesviruses such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV) establish life-long persistence in the vast majority of humans without showing any clinical symptoms. However, if the immune system is compromised, in for example, immunodeficient patients, they are at a higher risk for developing severe diseases, such as cancers, from these viruses [18]. In addition, small DNA viruses, such as human papillomaviruses and polyomaviruses, have coevolved with host species and are also frequently found in the human virome [19]. Although a small subset of these viruses encodes potent viral oncoproteins, the majority of persistent infections of these viruses are found in asymptomatic hosts. Unlike retroviruses, DNA viruses such as herpesviruses, papillomaviruses, and polyomaviruses normally maintain their genomes as multicopy episomes in persistently infected cells. These viral episomes, maintained in a covalently closed circular form, are assembled into chromatin with histones for autonomous replication [20]. However, these viruses also occasionally integrate into the host genome, despite no benefits for their replication from viral integration.

For these viruses to establish life-long persistence in the human virome, it is essential to avoid recognition and elimination by host antiviral immunity. Accordingly, almost all viruses in the human virome evade immune recognition by limiting viral replication and gene expression. The lack of viral gene expression and/or host immune tolerance leads to immune silence or an immune equilibrium state in the host. Thus, viral antigens from reactivated viral gene expression in tumor cells could be recognized as tumor neoantigens to induce antitumor immune responses.

How can endogenous retroviruses be reactivated to induce antitumor immunity?

Viral gene expression from HERVs are predominantly silenced by DNA methylation and histone modification, resulting in a transcriptionally inactive heterochromatic structure [21]. Thus, reactivating HERVs using epigenetic drugs has been proposed to trigger antitumor immune responses [22, 23].

DNA methyltransferase inhibitors

Reversing DNA methylation with DNA methyltransferase inhibitors (DNMTi), such as decitabine, reactivates expression of HERV RNA transcripts. Detection of these transcripts by the host double stranded RNA sensors, such as the toll-like receptors (TLRs) and melanoma differentiation-associated protein 5 (MDA5), drives an interferon (IFN) response [2, 24]. Due to chromosomal residence in the germline, HERV proteins should be generally considered as “self” peptides with their cognate T cells deleted in the thymus during T-cell development. However, due to the effective epigenetic silencing of HERVs, HERV-specific T cells are not removed during T-cell development and can be activated by transcriptional induction of HERVs [23]. Consequently, reactivated HERVs in tumor cells can be recognized by host as tumor neoantigens.

Indeed, reactivation of endogenous retroviruses (ERVs) by DNMTi enhances susceptibility to anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) ICI therapy in the B16 mouse melanoma model, a model that has been shown to be highly resistant to ICIs (Fig 1A) [2]. DNMTi treatment significantly up-regulates the expression of intercellular adhesion molecule 1 (ICAM1), interleukin-1 receptor antagonist (IL-1RA), CXCL10, CCL2, and CCL5 and increases the percentage of effector CD8+ T and natural killer (NK) cells in the TME [25]. Interestingly, these antitumor effects induced by DNMTi completely disappear when the IFN-α/β receptor α chain (IFNAR1) is blocked, suggesting that type I IFN signaling is critical. The combination therapy of DNMTi and histone deacetylase inhibitors (HDACi) synergistically increases reexpression of silenced ERVs in cancer cells and increases sensitivity to the anti-programmed cell death 1 (PD-1) therapy in lung [26] and ovarian cancers [25]. Interestingly, however, the HDACi treatment alone has little effect, suggesting that DNA demethylation is necessary for ERV reactivation.

thumbnail
Fig 1. Antiviral immune responses sensitize antitumor immunotherapy.

(A) Reactivation of ERVs by DNA demethylation induces an IFN response through cytosolic sensing of double stranded RNA. (B) Activation of virus-specific memory T cells induces antitumor CD8+ T cells through cross-presentation of viral peptides on DCs. CD8T, CD8+ T cells; CMV, cytomegalovirus; DC, dendritic cell; dsRNA, double stranded RNA; EBV, Epstein-Barr virus; ERV, endogenous retrovirus; Flu, influenza; IFN, interferon; MDA5, melanoma differentiation-associated protein 5; RIG-I, retinoic acid-inducible gene I.

https://doi.org/10.1371/journal.ppat.1008814.g001

Consistent with these findings in mouse models, the signatures of antitumor effector function and anti-PD-1 responsiveness are also positively associated with HERV expression in human cancers [27]. In addition, Haffner and colleagues found that type I IFN signaling and the number of CD8+ T cells are significantly higher in seminomatous testicular germ cell tumor (TGCT) characterized by constitutive DNA hypermethylation when compared to several non-seminomatous TGCTs that have normal methylation levels [28]. Beyond ERV reactivation, DNMTi also induces antitumor immune responses by activating transcription of tumor antigens (e.g., cancer-testis antigens) and Th1-type chemokines (e.g., CXCL9 and CXCL10) [6, 29]. These results suggest that DNMTi could be used as a useful multi-tool for cancer immunotherapy.

Cyclin-dependent kinase inhibitors

While it is well known that inhibitors of cyclin-dependent kinase (CDK) 4/6—the key regulators of the G1–S transition of the cell cycle—directly inhibit tumor growth by arresting cell cycle progression, recent studies have also discovered that CDK4/6 inhibitors can induce strong antitumor immune responses [3032]. CDK4/6 inhibitor treatment reactivates ERVs, produces type III IFNs, suppresses the proliferation of regulatory T cells, and down-regulates the expression of the E2F transcription factor target genes including DNA methyltransferase 1 (DNMT1) [30]. Interestingly, CDK4 also directly interacts with and stabilizes DNMT1, thus further supporting the notion that CDK4/6 inhibitors would impair DNMT1 activity and reactivate ERVs [33].

In addition to CDK4/6 inhibition, inhibition of CDK9—a transcriptional activator recruited to promote RNA polymerase II—also induces ERV expression and antitumor immune activation without DNA demethylation [4]. A selective CDK9 inhibitor blocks phosphorylation of the chromosome remodeling protein SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 (SMARCA4), which epigenetically silences a set of genes including ERVs independently of DNA methylation [4]. Accordingly, ERV reactivation by treatment of CDK4/6 or CDK9 inhibitors significantly enhances the response rates of the current ICI therapies.

How can quiescent antiviral immunity be reactivated to boost antitumor immunity?

The idea of using antiviral immunity for cancer treatment has been studied for over a century, and finally, the first oncolytic virus (Talimogene laherparepvec [T-VEC]) was approved by the Food and Drug Administration (FDA) in 2015 [34, 35]. Many different recombinant human viruses, including adenovirus, herpesvirus, poliovirus, reovirus, and poxvirus, are being used as oncolytic viruses in approximately 120 ongoing clinical trials to directly lyse tumor cells and indirectly stimulate antitumor immune responses (ClinicalTrials.gov) [36]. In addition to the oncolytic viruses, quiescent antiviral immune responses activated by viral peptides and vaccines can induce antitumor activity in the TME [5, 37].

Recent studies have shown that memory T cells specific to viruses in the human virome such as CMV and EBV are abundant in humans and can be activated by viral antigenic peptides [11, 37]. Although these host antiviral memory T-cell responses are not sufficient to eliminate life-long persistence of the viruses, treatment with viral peptides may be useful to awaken existing memory T-cell responses against exogenous viruses, such as CMV and EBV, persistent in the human virome to promote an antitumor response [37]. Accordingly, injection of CMV viral peptides significantly enhances the efficacy of the anti-PD-1 ligand 1 (PD-L1) therapy and suppresses tumor growth by activating an antiviral memory T-cell response (Fig 1B) [37].

The FDA-approved seasonal influenza vaccine greatly increases cross-presenting dendritic cells (DCs) and tumor antigen-specific CD8+ T cells in the TME (Fig 1B). These DCs and CD8+ T cells have been shown to play an important role in both antiviral and antitumor immune responses when the influenza vaccine is injected intratumorally [5]. Although adjuvants in vaccines normally boost immune responses against target pathogens, antitumor CD8+ T-cell responses are abrogated by adjuvant-enhanced regulatory B cells. These results suggest differential regulations of antiviral and antitumor immune responses despite their shared immune components and mechanisms. To further screen commercially available vaccines that activate the immunogenicity of cold tumors, Shekarian and colleagues tested 14 viral and bacterial vaccines and found that intratumoral injection of 2 rotavirus vaccines (Rotarix [GlaxoSmithKline Biologicals, Rixensart, Belgium] and RotaTeq [Merck, Kenilworth, NJ]) can activate antitumor immunity and aid in overcoming tumor resistance to ICI immunotherapies, suppressing tumor growth [38]. These results represent a potent mechanism to initiate key immune responses in tumors to assist in successful treatment.

Taken together, recent findings suggest that the awakening of antiviral host immune responses to the viruses in the human virome could be an effective force to fight against cancer.

References

  1. 1. Mokili JL, Rohwer F, Dutilh BE. Metagenomics and future perspectives in virus discovery. Curr Opin Virol. 2012;2(1):63–77. Epub 2012/03/24. pmid:22440968.
  2. 2. Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell. 2015;162(5):974–86. Epub 2015/09/01. pmid:26317466; PubMed Central PMCID: PMC4556003.
  3. 3. Zhao H, Ning S, Nolley R, Scicinski J, Oronsky B, Knox SJ, et al. The immunomodulatory anticancer agent, RRx-001, induces an interferon response through epigenetic induction of viral mimicry. Clin Epigenetics. 2017;9:4. Epub 2017/02/06. pmid:28149332; PubMed Central PMCID: PMC5270305.
  4. 4. Zhang H, Pandey S, Travers M, Sun H, Morton G, Madzo J, et al. Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell. 2018;175(5):1244–58 e26. Epub 2018/11/21. pmid:30454645; PubMed Central PMCID: PMC6247954.
  5. 5. Newman JH, Chesson CB, Herzog NL, Bommareddy PK, Aspromonte SM, Pepe R, et al. Intratumoral injection of the seasonal flu shot converts immunologically cold tumors to hot and serves as an immunotherapy for cancer. Proc Natl Acad Sci U S A. 2020;117(2):1119–28. Epub 2020/01/01. pmid:31888983; PubMed Central PMCID: PMC6969546.
  6. 6. Jones PA, Ohtani H, Chakravarthy A, De Carvalho DD. Epigenetic therapy in immune-oncology. Nat Rev Cancer. 2019;19(3):151–61. Epub 2019/02/07. pmid:30723290.
  7. 7. Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol. 2019;10:168. Epub 2019/02/26. pmid:30800125; PubMed Central PMCID: PMC6376112.
  8. 8. Gujar S, Pol JG, Kroemer G. Heating it up: Oncolytic viruses make tumors 'hot' and suitable for checkpoint blockade immunotherapies. Oncoimmunology. 2018;7(8):e1442169. Epub 2018/09/18. pmid:30221036; PubMed Central PMCID: PMC6136862.
  9. 9. Ng KW, Marshall EA, Bell JC, Lam WL. cGAS-STING and Cancer: Dichotomous Roles in Tumor Immunity and Development. Trends Immunol. 2018;39(1):44–54. Epub 2017/08/24. pmid:28830732.
  10. 10. Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017;7(3):264–76. Epub 2016/12/30. pmid:28031159; PubMed Central PMCID: PMC5733805.
  11. 11. Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, et al. Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature. 2018;557(7706):575–9. Epub 2018/05/18. pmid:29769722.
  12. 12. Virgin HW. The virome in mammalian physiology and disease. Cell. 2014;157(1):142–50. Epub 2014/04/01. pmid:24679532; PubMed Central PMCID: PMC3977141.
  13. 13. Moustafa A, Xie C, Kirkness E, Biggs W, Wong E, Turpaz Y, et al. The blood DNA virome in 8,000 humans. PLoS Pathog. 2017;13(3):e1006292. Epub 2017/03/23. pmid:28328962; PubMed Central PMCID: PMC5378407.
  14. 14. Bannert N, Kurth R. The evolutionary dynamics of human endogenous retroviral families. Annu Rev Genomics Hum Genet. 2006;7:149–73. Epub 2006/05/26. pmid:16722807.
  15. 15. Hohn O, Hanke K, Bannert N. HERV-K(HML-2), the Best Preserved Family of HERVs: Endogenization, Expression, and Implications in Health and Disease. Front Oncol. 2013;3:246. Epub 2013/09/26. pmid:24066280; PubMed Central PMCID: PMC3778440.
  16. 16. Garcia-Montojo M, Doucet-O'Hare T, Henderson L, Nath A. Human endogenous retrovirus-K (HML-2): a comprehensive review. Crit Rev Microbiol. 2018;44(6):715–38. Epub 2018/10/16. pmid:30318978; PubMed Central PMCID: PMC6342650.
  17. 17. Prusty BK, Krohne G, Rudel T. Reactivation of chromosomally integrated human herpesvirus-6 by telomeric circle formation. PLoS Genet. 2013;9(12):e1004033. Epub 2013/12/25. pmid:24367281; PubMed Central PMCID: PMC3868596.
  18. 18. Sehrawat S, Kumar D, Rouse BT. Herpesviruses: Harmonious Pathogens but Relevant Cofactors in Other Diseases? Front Cell Infect Microbiol. 2018;8:177. Epub 2018/06/12. pmid:29888215; PubMed Central PMCID: PMC5981231.
  19. 19. Chen X, Kost J, Sulovari A, Wong N, Liang WS, Cao J, et al. A virome-wide clonal integration analysis platform for discovering cancer viral etiology. Genome Res. 2019;29(5):819–30. Epub 2019/03/16. pmid:30872350; PubMed Central PMCID: PMC6499315.
  20. 20. Chiu YF, Sugden B. Plasmid Partitioning by Human Tumor Viruses. J Virol. 2018;92(9). Epub 2018/02/23. pmid:29467315; PubMed Central PMCID: PMC5899212.
  21. 21. Groh S, Schotta G. Silencing of endogenous retroviruses by heterochromatin. Cell Mol Life Sci. 2017;74(11):2055–65. Epub 2017/02/06. pmid:28160052.
  22. 22. Canadas I, Thummalapalli R, Kim JW, Kitajima S, Jenkins RW, Christensen CL, et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med. 2018;24(8):1143–50. Epub 2018/07/25. pmid:30038220; PubMed Central PMCID: PMC6082722.
  23. 23. Kassiotis G, Stoye JP. Immune responses to endogenous retroelements: taking the bad with the good. Nat Rev Immunol. 2016;16(4):207–19. Epub 2016/03/31. pmid:27026073.
  24. 24. Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell. 2015;162(5):961–73. Epub 2015/09/01. pmid:26317465; PubMed Central PMCID: PMC4843502.
  25. 25. Stone ML, Chiappinelli KB, Li H, Murphy LM, Travers ME, Topper MJ, et al. Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden. Proc Natl Acad Sci U S A. 2017;114(51):E10981–E90. Epub 2017/12/06. pmid:29203668; PubMed Central PMCID: PMC5754782.
  26. 26. Topper MJ, Vaz M, Chiappinelli KB, DeStefano Shields CE, Niknafs N, Yen RC, et al. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell. 2017;171(6):1284–300 e21. Epub 2017/12/02. pmid:29195073; PubMed Central PMCID: PMC5808406.
  27. 27. Smith CC, Beckermann KE, Bortone DS, De Cubas AA, Bixby LM, Lee SJ, et al. Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma. J Clin Invest. 2018;128(11):4804–20. Epub 2018/08/24. pmid:30137025; PubMed Central PMCID: PMC6205406.
  28. 28. Haffner MC, Taheri D, Luidy-Imada E, Palsgrove DN, Eich ML, Netto GJ, et al. Hypomethylation, endogenous retrovirus expression, and interferon signaling in testicular germ cell tumors. Proc Natl Acad Sci U S A. 2018;115(37):E8580–E2. Epub 2018/09/06. pmid:30181298; PubMed Central PMCID: PMC6140483.
  29. 29. Peng D, Kryczek I, Nagarsheth N, Zhao L, Wei S, Wang W, et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature. 2015;527(7577):249–53. pmid:26503055.
  30. 30. Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–5. Epub 2017/08/17. pmid:28813415; PubMed Central PMCID: PMC5570667.
  31. 31. Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018;8(2):216–33. Epub 2017/11/05. pmid:29101163; PubMed Central PMCID: PMC5809273.
  32. 32. Schaer DA, Beckmann RP, Dempsey JA, Huber L, Forest A, Amaladas N, et al. The CDK4/6 Inhibitor Abemaciclib Induces a T Cell Inflamed Tumor Microenvironment and Enhances the Efficacy of PD-L1 Checkpoint Blockade. Cell Rep. 2018;22(11):2978–94. Epub 2018/03/15. pmid:29539425.
  33. 33. Acevedo M, Vernier M, Mignacca L, Lessard F, Huot G, Moiseeva O, et al. A CDK4/6-Dependent Epigenetic Mechanism Protects Cancer Cells from PML-induced Senescence. Cancer Res. 2016;76(11):3252–64. Epub 2016/05/22. pmid:27206849.
  34. 34. Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007;15(4):651–9. Epub 2007/02/15. pmid:17299401.
  35. 35. Killock D. Skin cancer: T-VEC oncolytic viral therapy shows promise in melanoma. Nat Rev Clin Oncol. 2015;12(8):438. Epub 2015/06/17. pmid:26077044.
  36. 36. Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. Mol Ther Oncolytics. 2019;15:234–47. Epub 2019/12/25. pmid:31872046; PubMed Central PMCID: PMC6911943.
  37. 37. Rosato PC, Wijeyesinghe S, Stolley JM, Nelson CE, Davis RL, Manlove LS, et al. Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy. Nat Commun. 2019;10(1):567. Epub 2019/02/06. pmid:30718505; PubMed Central PMCID: PMC6362136.
  38. 38. Shekarian T, Sivado E, Jallas AC, Depil S, Kielbassa J, Janoueix-Lerosey I, et al. Repurposing rotavirus vaccines for intratumoral immunotherapy can overcome resistance to immune checkpoint blockade. Sci Transl Med. 2019;11(515). Epub 2019/10/28. pmid:31645452.