Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cancer Research Highlights

IFNγ, a Double-Edged Sword in Cancer Immunity and Metastasis

Chengfei Liu and Allen C. Gao
Chengfei Liu
Department of Urology, Comprehensive Cancer Center, University of California Davis, Sacramento, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Allen C. Gao
Department of Urology, Comprehensive Cancer Center, University of California Davis, Sacramento, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: acgao@ucdavis.edu
DOI: 10.1158/0008-5472.CAN-19-0083 Published March 2019
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

IFNγ has antitumorigenic effects; however, the findings of IFNγ in promoting the tumor cell survival and inducing adaptive immune resistance via CD4+ T-cell loss and programmed death ligand 1 (PD-L1) upregulation challenge this concept. Lo and colleagues determined that IFNγ induces epithelial–mesenchymal transition (EMT) by regulating the turnover of miRNA in prostate cancer, emphasizing the duplicitous effects of IFNγ. IFIT5, an IFN-induced tetratricopeptide repeat (IFIT) family member, was found to form a complex with the exoribonuclease-XRN1 to process miRNA maturation. These findings unveil a new IFNγ–STAT1–IFIT5–miRNA–EMT pathway in prostate cancer progression. The biphasic effects of IFNγ in prostate cancer raise concerns about its therapeutic application, which need to be evaluated in future studies.

See related article by Lo et al., p. 1098

In this issue of Cancer Research, Lo and colleagues (1) describe an interesting observation wherein IFNγ induces epithelial–mesenchymal transition (EMT) in prostate cancer cells by regulating the degradation of precursor miRNAs through a complex between the known IFNγ-stimulated RNA-binding protein, interferon-induced tetratricopeptide 5 (IFIT5), and the exoribonuclease candidate XRN1 (IFIT5-XRN1). Their study emerged from research on the GTPase-activating protein, DAB2IP, which has long been a focus for this group. They found that IFNγ promotes EMT in prostate cancer cells through DAB2IP, which has been previously identified as an upstream regulator of EMT, and went on to profile the miRNAs and show that miR-363 expression and maturation are specifically regulated by DAB2IP and that IFIT5 is a key factor regulating miR-363 turnover. The authors next sought to further characterize the network among IFIT5, miRNA, and EMT in prostate cancer. They first discovered that miR-363 suppresses EMT by targeting Slug in prostate cancer cells and that IFIT5 recognizes the unique 5′-end overhanging structure of pre-miR-363 to target it for degradation. Because IFIT5 does not possess ribonuclease activity, the authors then demonstrated that IFIT5 alone is not sufficient in regulating miRNA maturation and degradation but needs to form a complex with exoribonuclease-XRN1 to promote miR-363 turnover. To expand the overall effects of the network they established, the authors further examined other miRNAs that could be regulated by IFNγ. Two additional miRNAs, miR-101 and miR-128, were also identified to be involved in the IFIT5-EMT process. Finally, they found that IFIT5 is inversely correlated with miR-363, miR-101, and miR-128 and positively correlated with EMT markers ZEB1, Slug, and vimentin in prostate cancer specimens. From benchwork to clinical validation, Lo and colleagues identified a network of immune factors, transcriptional factors, and miRNAs involved in prostate cancer progression and metastasis.

Although we now recognize that miRNA plays crucial roles in prostate cancer progression, regulation of miRNA expression remains largely unknown. After transcription, pre-miRNA undergoes nuclear and cytoplasmic processing to become mature miRNA. Alteration of mature miRNA expression occurs in many different ways, such as SNP, miRNA tailing, editing, methylation, and regulation of stability (2). XRN1 is a 5′-3′ exoribonuclease that predominantly degrades miRNAs after they have been decapped in cells. Lo and colleagues discovered for the first time that IFIT5 recruits XRN1 to form a unique miRNA complex with the 5′-end of pre-miRNA molecules. The reciprocal correlation of IFIT5 and miR-363 and miR-101 expression in prostate cancer specimens further supports the role of IFNγ signaling in miRNA regulation. Human IFNγ is predominantly from infiltrating immune cells and whether this correlation also exists in these cells in prostate tumors needs to be investigated in future studies. Nevertheless, this function of IFIT5 in miRNA processing provides a new piece of evidence that IFNγ signaling regulates miRNA maturation and turnover in prostate cancer.

Over the last few decades, our understanding of the role of IFNγ in cancer immunity has been evolving. Numerous studies have reported that IFNγ is an important cytokine that facilitates both the innate and adaptive immune systems. Initial induction of IFNγ significantly suppresses tumor growth via immune activation; however, it can also induce CD4+ T-cell apoptosis, alter the CD4:CD8 ratio, and subsequently impair secondary antitumor immune responses (3). As a type II IFN, IFNγ plays both pro- and antitumorigenic roles in immunoediting, a process that consists of immunosurveillance and tumor progression (4). IFNγ plays a role in all three phases of the immunoediting process: elimination, equilibrium, and escape. During the elimination phase, natural killer (NK) cells, NK T cells, CD8+, and CD4+ T cells secrete IFNγ, which then activates macrophages, dendritic cells, Th1 CD4+ helper T cells, and B cells that lead to complete tumor elimination, suggesting that IFNγ is predominantly antitumorigenic in the tumor environment during this phase. During the equilibrium phase, the immune system is able to maintain immune-mediated tumor dormancy. This phase is poorly understood, in part, because of technical challenges in establishing mouse models, but it is known that IFNγ is required for maintaining tumor dormancy by inducing cancer senescence. Finally, during the escape phase, tumor cells grow and expand through mechanisms including adaptive and acquired immune resistance (5). Recent studies suggest that IFNγ can upregulate the expression of programmed-death ligand 1 (PD-L1), a membrane-bound immune inhibitory molecule on the surface of tumor cells. Because PD-L1 binds to programmed cell death protein 1 (PD1) expressed on the activated CD8+ T cells and leads to apoptotic T-cell death, IFNγ signaling activation facilities tumor cells escaping from the antitumor CD8+ T-cell cytotoxicity through formation of the immunosuppressive tumor microenvironment, promotes tumor cell survival, and induces adaptive resistance (6). Therefore, IFNγ possibly plays a protumorigenic role during the tumor immunity escape stage. Although application of immunotherapy in prostate cancer lags behind that in other cancer types, several clinical trials testing immune checkpoint inhibitors in patients with prostate cancer have begun (7). The exact role of IFNγ in prostate cancer immunity remains to be determined and the study by Lo and colleagues brings up more concern into the prostate cancer immunotherapy arena by reinforcing the dual nature of IFNγ in prostate cancer progression.

Metastasis is the primary underlying cause of fatality in patients with prostate cancer. Bone metastases occur in more than 90% of patients with advanced prostate cancer and are associated with poor survival. For men with metastatic prostate cancer, only one-third survive for 5 years after diagnosis (8). There is an urgent need to unravel potential resistant mechanisms that perpetuate disease progression during effective androgen receptor blockade and to devise ways of targeting resistant pathways. EMT is believed to be a crucial step in the conversion of early-stage disease to invasive and metastatic cancer (9). Immortalized human mammary epithelial cells acquire the mesenchymal phenotype and express stem cell markers after induction of EMT (10). The aberrant expression and localization of E-cadherin, N-cadherin, vimentin, Wnt5A, and ZEB1 appears to be important in prostate cancer invasion and bone metastasis. Although strong evidence supports EMT as the essential step in the progression and metastasis of prostate cancer, difficulties in identifying migratory cancer cells have precluded confirmation of the occurrence of EMT in vivo for many years. Thus, better understanding of upstream EMT regulators is urgent and important in the clinical arena. EMT can be triggered by tumor-associated fibroblasts, immune cells, and secreted soluble factors, such as Wnt ligands, TGFβ, EGF, and hepatocyte growth factor. These factors and inflammatory cytokines can exert their effects through autocrine or paracrine systems. Slug, Snail, ZEB1, ZEB2, and Twist have been previously identified as classical EMT regulators. In their study, Lo and colleagues report that IFNγ induces EMT in prostate cancer cells by regulating expression of EMT regulators and markers such as ZEB1, Slug, and vimentin as well as miR-363, miR-101, and miR-128. The take-home message from this comprehensive study is that the administration of IFNγ might not benefit patients with prostate cancer and possibly cause some harmful side effects. Nevertheless, clinical evidence and clinical trials would be required before this could be considered as a conclusion.

Together, IFNγ signaling is still largely unfathomable in prostate cancer. Considering the classical role of IFNγ in cancer immunoediting, the possibility of its utility in prostate cancer immunetherapy arena should not be disregarded. Prostate cancer immunotherapy awaits rigorous investigation to define the real targets and pathways involved in the therapy. A better grasp of the detailed mechanisms that underlie the effects of IFNγ in prostate cancer immunoediting appears to be necessary. Lo and colleagues have identified a novel tumor-promoting IFNγ–STAT1–IFIT5–miRNA–EMT pathway in prostate cancer cells, suggesting that IFNγ might serve as a master regulator in controlling several downstream signaling pathways, such as JAK-STAT1, IFIT5, DAB2IP, and miRNA signaling leading to prostate cancer progression and metastasis through EMT, raising concerns about its clinical application.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

This commentary did not receive any specific grant from any funding agency in the public, commercial, or not-for-profit sector.

  • Received January 18, 2019.
  • Accepted January 18, 2019.
  • Published first March 15, 2019.
  • ©2019 American Association for Cancer Research.

References

  1. 1.↵
    1. Lo UG,
    2. Pong RC,
    3. Yang D,
    4. Gandee L,
    5. Hernandez E,
    6. Dang A,
    7. et al.
    IFN-γ- induced IFIT5 promotes epithelial-to-mesenchymal transition in prostate cancer via miRNA processing. Cancer Res 2019;79:1098–112.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Ha M,
    2. Kim VN
    . Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 2014;15:509–24.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Berner V,
    2. Liu H,
    3. Zhou Q,
    4. Alderson KL,
    5. Sun K,
    6. Weiss JM,
    7. et al.
    IFN-gamma mediates CD4+ T-cell loss and impairs secondary antitumor responses after successful initial immunotherapy. Nat Med 2007;13:354–60.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Kaplan DH,
    2. Shankaran V,
    3. Dighe AS,
    4. Stockert E,
    5. Aguet M,
    6. Old LJ,
    7. et al.
    Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci U S A 1998;95:7556–61.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Alspach E,
    2. Lussier DM,
    3. Schreiber RD
    . Interferon gamma and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb Perspect Biol 2018 April 16 [Epub ahead of print].
  6. 6.↵
    1. Mandai M,
    2. Hamanishi J,
    3. Abiko K,
    4. Matsumura N,
    5. Baba T,
    6. Konishi I
    . Dual faces of IFNgamma in cancer progression: a role of PD-L1 induction in the determination of pro- and antitumor immunity. Clin Cancer Res 2016;22:2329–34.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Rescigno P,
    2. de Bono JS
    . Immunotherapy for lethal prostate cancer. Nat Rev Urol 2019;16:69–70.
    OpenUrl
  8. 8.↵
    1. Gundem G,
    2. Van Loo P,
    3. Kremeyer B,
    4. Alexandrov LB,
    5. Tubio JMC,
    6. Papaemmanuil E,
    7. et al.
    The evolutionary history of lethal metastatic prostate cancer. Nature 2015;520:353–7.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Thiery JP
    . Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2:442–54.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Mani SA,
    2. Guo W,
    3. Liao MJ,
    4. Eaton EN,
    5. Ayyanan A,
    6. Zhou AY,
    7. et al.
    The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133:704–15.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 79 (6)
March 2019
Volume 79, Issue 6
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Editorial Board (PDF)

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
IFNγ, a Double-Edged Sword in Cancer Immunity and Metastasis
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
IFNγ, a Double-Edged Sword in Cancer Immunity and Metastasis
Chengfei Liu and Allen C. Gao
Cancer Res March 15 2019 (79) (6) 1032-1033; DOI: 10.1158/0008-5472.CAN-19-0083

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
IFNγ, a Double-Edged Sword in Cancer Immunity and Metastasis
Chengfei Liu and Allen C. Gao
Cancer Res March 15 2019 (79) (6) 1032-1033; DOI: 10.1158/0008-5472.CAN-19-0083
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Disclosure of Potential Conflicts of Interest
    • Acknowledgments
    • References
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • The Key to BRCAness
  • Being Small and Intronic: miRNAs That Count
  • Adaptive Therapy and the Cost of Drug-Resistant Mutants
Show more Cancer Research Highlights
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement