Elsevier

DNA Repair

Volume 87, March 2020, 102802
DNA Repair

HPV induction of APOBEC3 enzymes mediate overall survival and response to cisplatin in head and neck cancer

https://doi.org/10.1016/j.dnarep.2020.102802Get rights and content

Highlights

  • HPV induces APOBEC3 expression in HNSC.

  • High APOBEC3 G expression correlates with better overall survival.

  • APOBEC3, Polβ and MSH6 knockdown results in cisplatin resistance.

  • Cisplatin resistance following A3 knockdown correlates with ICL repair.

Abstract

Human papillomavirus (HPV) is associated with the development of head and neck squamous cell carcinomas (HNSC). Cisplatin is used to treat HNSC and induces DNA adducts including interstrand crosslinks (ICLs). Previous reports have shown that HPV positive HNSC patients respond better to cisplatin therapy. Our previous reports highlight that loss of base excision repair (BER) and mismatch repair (MMR) results in cisplatin resistance. Of importance, uracil DNA glycosylase (UNG) is required to initiate the BER response to cisplatin treatment and maintain drug sensitivity. These previous results highlight that specific cytidine deaminases could play an important role in the cisplatin response by activating the BER pathway to mediate drug sensitivity. The APOBEC3 (A3) family of cytidine deaminases are enzymes that restrict HPV as part of the immune defense to viral infection. In this study, the Cancer Genome Atlas (TCGA) HNSC data were used to assess the association between the expression of the seven proteins in the A3 cytidine deaminase family, HPV-status and survival outcomes. Higher A3 G expression in HPV-positive tumors corresponds with better overall survival (OS) (HR 0.33, 95 % CI 0.11-0.93, p = 0.04). FaDu and Scc-25 HNSC cell lines were used to assess alterations in A3, BER and MMR expression in response to cisplatin. We demonstrate that A3, Polβ, and MSH6 knockdown in HNSC cells results in resistance to cisplatin and carboplatin as well as an increase in the rate of ICL removal in FaDu and Scc-25 HNSC cells. Our results suggest that A3s activate BER in HNSC, mediate repair of cisplatin ICLs and thereby, sensitize cells to cisplatin which likely contributes to the improved patient responses observed in HPV infected patients.

Introduction

Head and neck squamous cell carcinomas (HNSC) encompass cancers of the oral cavity, pharynx, larynx, nasal cavity, paranasal sinuses and salivary glands. The overall 5-year survival rate for oral and pharynx cancers is 65 %, but improves to 84 % for cancers diagnosed at a local stage [1]. Concurrent cisplatin and radiation, along with surgery, is the preferred regimen for the treatment of locally advanced HNSC [2].

Tobacco and alcohol use are common, modifiable risk factors associated with HNSC risk. Human papillomavirus (HPV) infection is associated with the development of several cancers including HNSCs [1,3,4]. Patients with HPV-positive tumors have better prognosis than HPV-negative tumors, due partly to an increase in sensitivity to chemotherapy and radiation therapy [[5], [6], [7], [8]]. Molecular differences between HPV-positive and -negative tumors include differences in immune cell subtypes and tumor metabolism [9]. The APOBEC3 (A3) family of proteins are antiviral cytidine deaminases that are part of the immune system, restricting HPV through the deamination of cytosines that leads to viral DNA mutations [[10], [11], [12], [13], [14], [15], [16]]. There are seven proteins in this family, denoted: A3A, A3B, A3C, A3D, A3F, A3G and A3H. Previous reports suggest that A3A and A3B are upregulated with HPV infection, either directly by E6 and E7, or through dysregulation of transcription [[17], [18], [19], [20]]. A3 expression is also induced by type 1 interferons (IFN) [[21], [22], [23], [24]]. Off-target A3 deamination leads to a specific mutational signature enriched in HPV-positive cases [25,26]. This mutational signature is also found in other cancer types and thought to be part of tumor development [[27], [28], [29]]. The A3 mutational signature has been shown to be associated with HPV-positive HNSC cases in TCGA data [26].

Cisplatin is part of standard treatment of HNSC, either in combination with radiotherapy or adjuvant therapy following surgery [2]. A clinical trial detailed by Ang et al. showed that HPV status was a strong prognostic indicator of survival of oropharyngeal cancer patients treated with cisplatin in combination with standard or accelerated-fractionation radiotherapy [30]. Current research is focusing on decreasing treatment doses in HPV-positive patients to decrease side effects, as HPV-positive cases tend to have better survival and response to therapy [[5], [6], [7], [8]]. While both HPV-positive and –negative patients respond to cisplatin, there are significant disparities in the responses between HPV-status groups [31]. A recent study found that HPV-negative patients obtain a survival benefit with higher doses of cisplatin, whereas HPV-positive patients have survival benefit at lower doses [32]. Platinum agents, including cisplatin, form DNA adducts that lead to apoptosis if not repaired. Cisplatin and carboplatin form monoadducts, intrastrand adducts and interstrand crosslinks (ICLs). ICLs form at dGpC sites and are covalently linked between the guanines on opposing strands of DNA. We have previously shown in several cancer cell types that base excision repair (BER) and mismatch repair (MMR) proteins sensitize cells to cisplatin and carboplatin by preventing the removal of ICLs [[33], [34], [35], [36]]. BER and MMR physically prevent nucleotide excision repair and homologous recombination from removing ICLs by non-productively processing DNA base damage adjacent to these ICLs. Cisplatin and carboplatin ICLs distort the DNA helix that forces the cytosines that were bonded to the guanines to be extrahelical [37]. This specific structure may be opportune for enzymatic deamination of the extrahelical cytosines, resulting in uracils and thus forming substrates for BER activation. Of importance, we have previously shown that uracil DNA glycosylase (UNG) is required for subsequent BER processing to mediate cisplatin and carboplatin sensitivity as well as inhibiting ICL DNA repair [33].

Due to the induction and subsequent deamination activity of A3 enzymes in HPV infection and better survival in HPV-positive patients, we investigated whether expression of A3s alters survival in HNSC and alter cisplatin and carboplatin sensitivity. Since A3 expression is increased with HPV infection, we propose that this increase in A3 expression may be a factor in the better survival of HPV-positive patients following standard treatment with cisplatin. We also tested whether A3s sensitize HNSC cells to cisplatin and carboplatin, as we hypothesize that their deamination activity activates BER and MMR, ultimately altering ICL DNA repair.

Section snippets

Chemicals

Cisplatin, oxaliplatin, and carboplatin were purchased from Sigma-Aldrich. For preparation, cisplatin, carboplatin and oxaliplatin were diluted in 1X PBS for a stock concentration of 1 mM and vortexed until drug was completely dissolved followed by filtration through 0.2 μm filters. Cisplatin and carboplatin were prepared fresh before each experiment. Oxaliplatin was stored at −80 °C and used within six months.

Cell lines

The human pharynx squamous cell carcinoma FaDu cells and the human tongue squamous

Results

We initially compared the expression of A3s by HPV-positive or –negative status in HNSC. A3B, A3C, A3D, A3 F, A3 G, and A3H had significantly higher mean expression in HPV-positive tumors than HPV-negative tumors (p-values <1e−11, Fig. 1 and supplemental table 1), but A3A did not.

Univariate analysis through Kaplan-Meier survival curves showed no statistically significant differences in OS in HNSC cases by A3 expression dichotomized by the median, except A3A (Supplemental Fig. 1). Patients with

Discussion

A3s are cytidine deaminases involved in host-cell immunity to target viral DNA and protect the cell against viral infection, including HPV. We found HPV-positive HNSC cases more highly express A3s, except for A3A, compared to HPV-negative HNSC, which is consistent with an upregulation of A3s due to HPV infection. The impact of A3s on OS differed by HPV status, with HPV-positive tumors with high expression of A3 G having better OS, while HPV-negative tumors with high A3 F expression had worse

Author contributions

KLC, ANS, MLC and SMP conceived the project. KLC and ANS did the TCGA analysis. KLC and EE conducted the biological experiments. SK and JJR assisted with patient analysis. SK assisted with statistical analyses. KLC and ANS wrote the initial draft of the manuscript. MLC and SMP oversaw the project.

Declaration of Competing Interest

The authors declare there are no conflicts of interest.

Acknowledgments

This work was supported in part by the National Institute of Health Ruth L. Kirschstein National Research Service Award (T32-CA00953 to KLC, 1F31CA22133301 to ANS); Susan G. Komen for the Cure (GTDR14299438 to MLC, ANS, and KLC); and the National Institutes of Health (R01CA229535) awarded to SMP. The authors thank the members of the Patrick lab for carefully reading the manuscript. The results shown here are generated from TCGA (https://cancergenome.nih.gov).

References (56)

  • A. Kothandapani et al.

    Novel Role of base excision repair in mediating cisplatin cytotoxicity

    J. Biol. Chem.

    (2011)
  • A. Sawant et al.

    Differential role of base excision repair proteins in mediating cisplatin cytotoxicity

    DNA repair

    (2017)
  • A. Sawant et al.

    Role of mismatch repair proteins in the processing of cisplatin interstrand cross-links

    DNA repair

    (2015)
  • R.L. Siegel et al.

    CA Cancer J. Clin.

    (2019)
  • A.D. Colevas et al.

    NCCN guidelines insights: head and neck cancers, version 1.2018

    J. Natl. Compr. Cancer Netw.

    (2018)
  • S. Marur et al.

    HPV-associated head and neck Cancer: a virus-related Cancer Epidemic - a review of epidemiology, biology, virus detection and issues in management

    Lancet Oncol.

    (2010)
  • T. Ramqvist et al.

    An epidemic of oropharyngeal squamous cell carcinoma (OSCC) due to human papillomavirus (HPV) infection and aspects of treatment and prevention

    Anticancer Res.

    (2011)
  • S. Marur et al.

    Head and neck Cancer: changing epidemiology, diagnosis, and treatment

    Mayo Clin. Proc.

    (2008)
  • L. Vidal et al.

    Human papillomavirus in HNSCC: recognition of a distinct disease type

    Hematol. Oncol. Clin. North Am.

    (2008)
  • S.J. Smeets et al.

    Genetic Classification of Oral and Oropharyngeal Carcinomas Identifies Subgroups with a Different Prognosis

    Cell. Oncol. Official J. Inte. Soc. Cell. Oncol.

    (2009)
  • K. Lindel et al.

    Human papillomavirus positive squamous cell carcinoma of the oropharynx

    Cancer

    (2001)
  • R. Krupar et al.

    Immunologic and metabolic characteristics of HPV-negative and HPV-positive head and neck squamous cell carcinomas are strikingly different

    Virchows Arch.

    (2014)
  • A.M. Sheehy et al.

    Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein

    Nature

    (2002)
  • H. Zhang et al.

    The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA

    Nature

    (2003)
  • B. Mangeat et al.

    Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts

    Nature

    (2003)
  • R. Harris et al.

    DNA deamination mediates innate immunity to retroviral infection

    Cell

    (2003)
  • A.J. Schumacher et al.

    APOBEC3G hypermutates genomic DNA and inhibits Ty1 retrotransposition in yeast

    Proc. Nat. Academy Scie. U. S. A.

    (2005)
  • C. Esnault et al.

    APOBEC3G cytidine deaminase inhibits retrotransposition of endogenous retroviruses

    Nature

    (2005)
  • J.A. Dutko et al.

    Inhibition of a yeast LTR retrotransposon by human APOBEC3 cytidine deaminases

    Curr. Biol.

    (2005)
  • C.J. Warren et al.

    APOBEC3A functions as a restriction factor of human papillomavirus

    J. Virol.

    (2015)
  • V.C. Vieira et al.

    Human papillomavirus E6 triggers upregulation of the antiviral and Cancer Genomic DNA deaminase APOBEC3B

    mBio

    (2014)
  • S. Mori et al.

    Identification of APOBEC3B promoter elements responsible for activation by human papillomavirus type 16 E6

    Biochem. Biophycial. Res. Commun.

    (2015)
  • M. Periyasamy et al.

    p53 controls expression of the DNA deaminase APOBEC3B to limit its potential mutagenic activity in cancer cells

    Nucleic Acids Res.

    (2017)
  • H. Mehta et al.

    IFN-alpha and lipopolysaccharide upregulate APOBEC3 mRNA through different signaling pathways

    J. Immunol.

    (2012)
  • Y. Li et al.

    IFN-alpha mediated base excision repair pathway correlations with antiviral response against hepatitis B virus infection

    Sci. Rep.

    (2017)
  • M. Bonvin et al.

    Interferon-inducible expression of APOBEC3 editing enzymes in human hepatocytes and inhibition of hepatitis B virus replication

    Hepatology

    (2006)
  • G. Peng et al.

    Induction of APOBEC3 family proteins, a defensive maneuver underlying interferon-induced anti-HIV-1 activity

    J. Exp. Med.

    (2006)
  • M.B. Burns et al.

    Evidence for APOBEC3B mutagenesis in multiple human cancers

    Nat. Genet.

    (2013)
  • 1

    These authors Equally Contributed to this work.

    2

    These authors share senior authorship.

    View full text