1,718
Views
0
CrossRef citations to date
0
Altmetric
Author’s Views

APOBEC3 as a driver of genetic intratumor heterogeneity

ORCID Icon, , , , , & show all
Article: 2014734 | Received 03 Nov 2021, Accepted 01 Dec 2021, Published online: 03 Jan 2022

ABSTRACT

Our recent study revealed that APOBEC3B is upregulated during the preinvasive stages of non-small cell lung cancer and breast cancer. In addition to its role in mediating single nucleotide variants, we propose that APOBEC3 promotes copy number intratumor heterogeneity prior to invasion, providing a substrate for cancer evolution.

Analysis of sequencing data from tumor samples has revealed extensive intratumor heterogeneity at the single nucleotide as well as copy number level. Genomic intratumor heterogeneity is clinically relevant since it has been linked to immune evasion, anti-cancer treatment resistance and disease progression. Therefore, identifying drivers of intratumor heterogeneity could provide targets to intercept treatment resistance, disease progression and potentially even cancer initiation in high-risk patients.

Mutational signature analysis of sequencing data has revealed molecular underpinnings of mutagenic processes in cancer. One of the most pervasive signatures among different cancer types is an APOBEC3-mediated mutational signature. Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3; A3) is a family of cytidine deaminases consisting of 7 genes which are a part of the innate immune system. APOBEC3 forms a barrier against viral and transposon replication through cytidine deaminase-dependent and independent mechanisms. APOBEC3-mediated mutagenesis in viral, transposon and genomic DNA occurs through deamination of cytidines present in single-stranded DNA into uracils. Uracil can either be reverted back into a cytosine by the DNA damage repair system or be replaced by a thymine or guanine by the DNA replication machinery creating a single nucleotide mutation. This APOBEC3 mutational signature consists of C-to-T and C-to-G mutations in TCA and TCT trinucleotide motifs. The APOBEC3 family members A3A and A3B appear to be the main drivers of this signature, with the former appearing to be a more dominant mutator.Citation1 Our group and others have shown that APOBEC3 is a major driver of subclonal mutagenesis and cell-to-cell variation within several cancer types.Citation2

APOBEC3 activity occurs in preinvasive lesions and cancer

In a recent study,Citation3 we investigated APOBEC3 expression in non-small cell lung cancer (NSCLC) development and found extensive nuclear immunohistochemical staining for A3A and A3B in preinvasive lesions of lung adenocarcinoma and lung squamous cell carcinoma, but absent in normal epithelial cells. We confirmed these observations in independent gene expression datasets of NSCLC, revealing that preinvasive samples with moderate dysplasia had transcriptional upregulation of different APOBEC3 genes, including A3A and A3B, relative to normal epithelium. Besides NSCLC, we confirmed an increase in A3B gene expression in preinvasive breast lesions. In addition, whole exome sequencing of matched preinvasive and invasive lesions in 2 NSCLC patients revealed that APOBEC3-mediated subclonal mutagenesis can occur at the preinvasive stage prior to malignant transformation.

A3B induces chromosomal instability before malignant transformation

We identified that A3B-expressing cells exhibited a slower DNA replication fork speed relative to isogenic A3B knockout cells, supporting data from other groups,Citation4 collectively suggesting that A3B contributes to replication stress. Isogenic A3B wild type and A3B knockout cell lines were studied throughout different cell cycle phases. A3B wild type cells contained more Fanconi anemia complementation group D2 (FANCD2) foci in prometaphase, more metaphase breaks at the fragile histidine triad diadenosine triphosphatase (FHIT) common fragile site locus, more FANCD2-flanked ultrafine bridges and more tumor protein 53-binding protein 1 (TP53BP1, best known as 53BP1) nuclear bodies in the G1 cell cycle phase relative to A3B knockout cells. These observations suggest that A3B-mediated replication stress promotes double-stranded DNA breaks at late replicating common fragile sites. Interestingly, these phenotypes manifested only upon the addition of low-dose aphidicolin or the expression of a RAS oncogene, suggesting A3B might need some level of replication stress in order to induce genome instability. A3B wild type cells displayed higher frequencies of chromosome missegregation relative to A3B knockout cells. Furthermore, within an A3B-inducible mouse model of lung cancer, A3B-overexpressing cells contained higher levels of phosphorylated replication protein A (RPA) and higher frequencies of chromosome missegregation relative to A3B-deficient cells, indicative of replication stress and chromosomal instability, respectively. Besides cytidine deamination on ssDNA, recent studies have also implicated APOBEC3 in chromatin modification.Citation5 It is therefore conceivable that APOBEC3 can promote genome instability through distinct but not mutually exclusive mechanisms. Depending on the cell line and cancer type, other APOBEC3 family members such as A3A have recently also been shown to reduce DNA replication fork speed and to be involved in promoting chromosomal instability.Citation6,Citation7

APOBEC3 as a therapeutic target in cancer

Taken together, these findings are of interest since in NSCLC, chromosomal instability in the context of somatic DNA copy number heterogeneity has been associated with an increased risk of recurrence or death (see ).Citation8 With APOBEC3 family members being drivers of single nucleotide variants and chromosomal instability, it might be attractive to inhibit relevant APOBEC3 family members. Combination therapy with APOBEC3 inhibitors might be able to impede tumor evolution and the acquisition of treatment-resistant clones. A more controversial approach might seek to hyperactivate relevant APOBEC3 family members, beyond an optimal range in which genome instability increases cancer cell fitness, to drive cell autonomous lethality due to excessive genome instability. Recent evidence suggests that A3B overexpression and ensuing mutagenesis increases sensitivity to immunotherapies,Citation9 although there are also concerns that subclonal neoantigen heterogeneity may fuel an immune suppressive microenvironment and immune evasion.Citation10 The timing of APOBEC3 induction and the ensuing clonal or subclonal repertoire of neoantigens may determine sensitization to immune-mediated predation. In summary, our experimental and clinical data suggest that A3B fuels chromosomal instability in the context of somatic copy number diversity in addition to single nucleotide heterogeneity in early NSCLC development, enabling downstream selection and cancer evolution. In the future it will be of interest to address whether eradicating genomically unstable preinvasive cancer cells, or inhibiting APOBEC3 in carcinoma in situ, can prevent progression into invasive cancer.

Figure 1. Upregulation of A3B levels in preinvasive cancer promotes genome instability.

A, Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3; A3) gene expression increases during early non-small cell lung cancer (NSCLC) evolution. A3B protein levels are especially high in carcinoma in situ samples relative to normal lung epithelium. B and C, example of cells that have low and high A3B levels, respectively. A3B promotes replication stress and the accumulation of under-replicated DNA, leading to the formation of ultrafine bridges, chromosome missegregation and tumor protein 53-binding protein 1 (TP53BP1, best known as 53BP1) nuclear bodies.
Figure 1. Upregulation of A3B levels in preinvasive cancer promotes genome instability.

Acknowledgments

We apologize to our colleagues for the omission of many important contributions to the field, and their references, due to space limitations.

Disclosure statement

S.F.B. holds a patent related to some of the work described targeting CIN and the cGAS-STING pathway in advanced cancer. He owns equity in, receives compensation from, and serves as a consultant and the Scientific Advisory Board and Board of Directors of Volastra Therapeutics Inc. C.S. acknowledges grant support from Pfizer, AstraZeneca, Bristol Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Invitae (previously Archer Dx Inc) - collaboration in minimal residual disease sequencing technologies, and Ono Pharmaceutical, and is an AstraZeneca Advisory Board member and Chief Investigator for the MeRmaiD1 clinical trial. C.S has consulted for Amgen, Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol Myers Squibb, Celgene, Astra Zeneca, Illumina, Genentech, Roche-Ventana, GRAIL, Medicxi, Metabomed, Bicycle Therapeutics, Roche Innovation Centre Shanghai, and the Sarah Cannon Research Institute, C.S. has stock options in Apogen Biotechnologies, Epic Bioscience, GRAIL, and has stock options and is co-founder of Achilles Therapeutics. Patents: C.S. holds European patents relating to assay technology to detect tumour recurrence (PCT/GB2017/053289); to targeting neoantigens (PCT/EP2016/059401), identifying patent response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), identifying patients who respond to cancer treatment (PCT/GB2018/051912), a US patent relating to detecting tumour mutations (PCT/US2017/28013) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892). No potential conflicts of interest were disclosed by the other authors.

Additional information

Funding

S.F.B. is supported by the Office of the Director, the NIH under Award Number National Institutes of Health DP5OD026395 High-Risk High-Reward Program, the NCI Breast Cancer SPORE (National Cancer Institute P50CA247749) and R01 (R01CA256188-01), the Burroughs Wellcome Fund Career Award for Medical Scientists, the Parker Institute for Immunotherapy at MSKCC, the Josie Robertson Foundation, and the MSKCC core grant P30-CA008748. J.B. and his team were funded by grants from the Danish Cancer Society (R1123-A7785-15-S2 and R167-A11068), the Novo Nordisk Fonden (16854 and Novo Nordisk Fonden 0060590), the Lundbeck foundation (Lundbeckfonden R266-2017-4289 and R322-2019-2577), the Swedish Research council (Swedish Research Council VR-MH 2014-46602-117891-30), The Swedish Cancer Foundation/Cancerfonden (The Swedish Cancer Foundation 170176), and the Danish national research foundation(project CARD, DNRF 125). N.K. receives funding from Cancer Research UK.C.S. is Royal Society Napier Research Professor (RP150154). This work was supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (FC001169), the UK Medical Research Council FC001169, and the Wellcome Trust (FC001169). This research was funded in whole, or in part, by the Wellcome Trust (FC001169). For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission. C.S. is funded by Cancer Research UK (TRACERx, PEACE and CRUK Cancer Immunotherapy Catalyst Network), Cancer Research UK Lung Cancer Centre of Excellence (C11496/A30025), the Rosetrees Trust, Butterfield and Stoneygate Trusts, NovoNordisk Foundation (ID16584), Royal Society Professorship Enhancement Award (RP/EA/180007), the National Institute for Health Research (NIHR) Biomedical Research Centre at University College London Hospitals, the Cancer Research UK-University College London Centre, Experimental Cancer Medicine Centre, and the Breast Cancer Research Foundation (The Breast Cancer Research Foundation BCRF 20-157). This work was supported by a Stand Up To Cancer‐LUNGevity-American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT23-17 to S.M. Dubinett and A.E. Spira). Stand Up To Cancer is a division of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the Scientific Partner of SU2C. CS is in receipt of an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. H2020 European Research Council 835297);The Rosetrees Trust;The National Institute for Health Research (NIHR) Biomedical Research Centre at University College London Hospitals;The Cancer Research UK-University College London Centre;Parker Institute for Cancer Immunotherapy.

References

  • Jalili P, Bowen D, Langenbucher A, Park S, Aguirre K, Corcoran RB, Fleischman AG, Lawrence MS, Zou L, Buisson R, et al. Quantification of ongoing APOBEC3A activity in tumor cells by monitoring RNA editing at hotspots. Nat Commun. 2020;11(1):2971. doi:10.1038/s41467-020-16802-8.
  • Swanton C, McGranahan N, Starrett GJ, Harris RS. APOBEC enzymes: mutagenic fuel for cancer evolution and heterogeneity. Cancer Discov. 2015;5:704–4. doi:10.1158/2159-8290.CD-15-0344.
  • Venkatesan S, Angelova M, Puttick C, Zhai H, Caswell DR, Lu W-T, Dietzen M, Galanos P, Evangelou K, Bellelli R, et al. Induction of APOBEC3 exacerbates DNA replication stress and chromosomal instability in early breast and lung cancer evolution. Cancer Discov. 2021;11(10):2456–2473. doi:10.1158/2159-8290.CD-20-0725.
  • Nikkila J, Kumar R, Campbell J, Brandsma I, Pemberton HN, Wallberg F, Nagy K, Scheer I, Vertessy BG, Serebrenik AA, et al. Elevated APOBEC3B expression drives a kataegic-like mutation signature and replication stress-related therapeutic vulnerabilities in p53-defective cells. Br J Cancer. 2017;117(1):113–123. doi:10.1038/bjc.2017.133.
  • Wang D, Li X, Li J, Lu Y, Zhao S, Tang X, Chen X, Li J, Zheng Y, Li S, et al. APOBEC3B interaction with PRC2 modulates microenvironment to promote HCC progression. Gut. 2019;68(10):1846–1857. gutjnl-2018-317601. doi:10.1136/gutjnl-2018-317601.
  • Wörmann SM, Zhang A, Thege FI, Cowan RW, Rupani DN, Wang R, Manning SL, Gates C, Wu W, Levin-Klein R, et al. APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis. Nat Cancer. 2021. doi:10.1038/s43018-021-00268-8.
  • Mehta KPM, Lovejoy CA, Zhao R, Heintzman DR, Cortez D. HMCES maintains replication fork progression and prevents double-strand breaks in response to APOBEC deamination and abasic site formation. Cell Rep. 2020;31:107705. doi:10.1016/j.celrep.2020.107705.
  • Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, Shafi S, Johnson DH, Mitter R, Rosenthal R, et al. Tracking the evolution of non–small-cell lung cancer. N Engl J Med. 2017;376(22):2109–2121. doi:10.1056/NEJMoa1616288.
  • Driscoll CB, Schuelke MR, Kottke T, Thompson JM, Wongthida P, Tonne JM, Huff AL, Miller A, Shim KG, Molan A. APOBEC3B-mediated corruption of the tumor cell immunopeptidome induces heteroclitic neoepitopes for cancer immunotherapy. Nat Commun. 2020;11(790). doi:10.1038/s41467-020-14568-7
  • Wolf Y, Bartok O, Patkar S, Eli GB, Cohen S, Litchfield K, Levy R, Jiménez-Sánchez A, Trabish S, Lee JS, et al. UVB-Induced tumor heterogeneity diminishes immune response in melanoma. Cell. 2019;179(1):219–235 e221. doi:10.1016/j.cell.2019.08.032.