Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 1.
Published in final edited form as: Nat Genet. 2022 Oct 24;54(11):1599–1608. doi: 10.1038/s41588-022-01196-8

Addressing the benefits of inhibiting APOBEC3-dependent mutagenesis in cancer

Mia Petljak 1,*,**, Abby M Green 2,3,*, John Maciejowski 4,*, Matthew D Weitzman 5,6,7,*
PMCID: PMC9700387  NIHMSID: NIHMS1843747  PMID: 36280735

Abstract

Mutational signatures associated with APOBEC3 cytosine deaminase activity have been found in over half of cancer types, including some therapy resistant and metastatic tumors. Driver mutations can occur in APOBEC3-favored sequence contexts, suggesting that mutagenesis by APOBEC3 enzymes may drive cancer evolution. The APOBEC3-mediated signatures are often detected in subclonal branches of tumor phylogenies and are acquired in cancer cell lines over long periods of time, indicating that APOBEC3 mutagenesis can be ongoing in cancer. Collectively, these and other observations have led to the proposal that APOBEC3 mutagenesis represents a disease-modifying process that could be inhibited to limit tumor heterogeneity, metastasis and drug resistance. However, critical aspects of APOBEC3 biology in cancer and in healthy tissues have not been clearly defined, limiting well-grounded predictions regarding the benefits of inhibiting APOBEC3 mutagenesis in different settings in cancer. We discuss the relevant mechanistic gaps, and strategies to address them, to investigate whether inhibiting APOBEC3 mutagenesis may confer clinical benefits in cancer.

Editor summary:

This Perspective addresses next steps to investigate the predictions that inhibition of APOBEC3-mediated mutagenesis may limit tumor heterogeneity, metastasis and drug resistance in a broad range of cancer types, by highlighting gaps in our understanding of APOBEC3 biology in cancer and in healthy tissues, and strategies to address them.

Introduction

The apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) family of cytosine deaminases restrict viral pathogenesis through base-editing of viral single-stranded (ss) DNA and RNA molecules1. The APOBEC enzymes emerged as a source of a frequent mutational pattern in cancer genomes, characterized by mutations at cytosine bases in TCN sequence contexts (mutated base is underlined, N is any base)2-5. Mathematical deconvolution of signatures of individual mutational processes in cancer led to identification of single base substitution (SBS) signatures of non-clustered (termed SBS2 and SBS13) and clustered (kataegis, omikli, kyklonas) cytosine mutations in APOBEC-preferred sequence contexts, thus improving the quantification of APOBEC-associated mutations in individual cancers6-10. APOBEC-associated signatures have been found in approximately 40 - 70% of cancer genomes, with a particular prominence in cancers of breast, bladder, lung, cervix and esophagus, suggesting that APOBEC mutagenesis represents one of the most prevalent mutational processes in cancer7,9,11,12. Multiple associations indicate that members of the APOBEC3 subfamily may represent the major origins of APOBEC-associated in most cancer types3-5,13. Six out of seven members of the APOBEC3 subfamily preferentially deaminate cytosine bases in TCN trinucleotides that are commonly mutated in cancer14,15 (Fig. 1A). In line with these associations, deletion of endogenous APOBEC3s from human cancer cell lines severely decreases the accumulation of APOBEC-associated mutations2.

Figure 1. APOBEC3 mutagenesis is predicted to be a disease-modifying process in cancer that may be exploited therapeutically.

Figure 1.

A) APOBEC3 enzymes convert cytosine to uracil by deamination of cytosine bases in single-stranded DNA. Depending on the subsequent uracil-processing, different types of mutations can arise. Note, translesion synthesis (TLS) is predicted to give rise to multiple mutation types (C>A, C>G, C>T) at cytosine bases. UNG (uracil DNA glycosylase). B) (1) APOBEC3 mutagenesis is speculated to drive cancer cell evolution and associated phenotypes, including tumor heterogeneity, therapeutic resistance and metastases. (2) Inhibition of APOBEC3 mutagenesis alongside the standards of care may limit phenotypes associated with cancer cell evolution. The extent to which such phenotypes would be limited upon APOBEC3 inhibition in individual cancers likely depends in part on the strength of other mutational processes that contribute to cancer evolution. (3) If mutations driving resistance to a standard of care have already been acquired in a cancer before the start of the treatment, APOBEC3 inhibition alongside the standard of care may diminish evolution of tumor heterogeneity, but is not predicted to eliminate resistance to the relevant therapy.

Despite progress, the proposal that mutagenesis by APOBEC3 enzymes represents a disease-modifying process that could be inhibited to confer clinical benefit in various settings in cancer16-28 (Fig. 1B) largely rely on poorly defined roles of APOBEC3 deaminases in cancer and in healthy tissues. First, available surrogate measures of APOBEC3-mediated mutagenesis in cancer produce conflicting predictions regarding the relative mutagenic contributions of individual APOBEC3 enzymes3-5,29,30. Second, causal links between mutagenesis by individual APOBEC3 enzymes and features of cancer evolution, including tumor heterogeneity, therapy resistance and metastasis, are scarce. Third, the hypothesis that restriction of APOBEC3 mutagenesis may diminish phenotypes associated with cancer evolution awaits thorough investigations across different cancer types and therapy settings where such predictions have been made. Fourth, the therapeutic windows during which APOBEC3 inhibition may confer clinical benefit have not been defined. Fifth, the mechanisms by which APOBEC3 enzymes are regulated in healthy tissues or become dysregulated in cancer are largely unknown, which limits the scope and precision of potential therapeutic opportunities. Finally, the endogenous functions of APOBEC3 enzymes are incompletely understood, which complicates predictions regarding potential toxicities from modulating APOBEC3 activities.

Progress has been compromised by the limited availability of readouts of active mutagenesis by individual APOBEC3 enzymes and limited investigations in preclinical models that closely recapitulate physiological contexts in which APOBEC3 mutagenesis is active. Here, we lay out criteria and strategies to define specific readouts of mutagenesis by individual APOBEC3 enzymes and to investigate impact of mutagenesis by individual enzymes on cancer evolution. We discuss the need to systematically characterize mechanisms of APOBEC3 misregulation in cancer and APOBEC3 functions in healthy tissues. Addressing these gaps is critical to realize the potential benefits of inhibiting APOBEC3 mutagenesis in cancer.

Addressing the readouts of APOBEC3 mutagenesis

APOBEC3 mutagenesis in cancer genomes is inferred by quantifying mutations at APOBEC3-associated sequence contexts9,10,31-34, measuring APOBEC3 transcript and protein levels22,23,34,35, in vitro APOBEC3 DNA deamination assays22,35,36 and a 3D-PCR-based APOBEC3A-associated RNA editing assay37 (Fig. 2). Aside from measuring expression of individual APOBEC3 enzymes and APOBEC3A-associated RNA editing, these readouts have not been shown to be specific to individual APOBEC3 enzymes in human cancer cells. Furthermore, mutations detected in cancer genomes represent imprints of historically incurred APOBEC3 mutagenesis, which may not always be ongoing38, while other readouts have not been directly connected to active genomic mutagenesis by APOBEC3 enzymes. Thus, readouts of active mutagenesis by individual APOBEC3 enzymes are not clearly defined.

Figure 2. Surrogate readouts of active mutagenesis by individual APOBEC3 enzymes.

Figure 2.

The degree to which assays of APOBEC3 expression (mRNA and protein) and deaminase activity upon DNA probes (e.g. probe cleavage assays) or upon endogenous RNA (e.g. 3D-PCR of commonly targeted transcripts) reflect active mutagenesis upon a genome is not clear. Mutations in cancer (including mutational signatures, mutations at extended sequence contexts and mutations at hairpin loops) indicate the historic exposure of a genome to APOBEC3 mutagenesis, but such readouts do not inform on whether APOBEC3 mutagenesis is active in a given tumor at the time of sampling. Pol, polymerase; A3A, APOBEC3A; A3B, APOBEC3B; RTCN/YTCN: R, purine base; Y, pyrimidine; N, any base.

The limitations inherent to the surrogate readouts of active mutagenesis by APOBEC3 enzymes are illustrated in often inconclusive or discordant results regarding the relative contributions of individual APOBEC3 enzymes to mutations in cancer. Based on multiple associations, APOBEC3A and APOBEC3B have emerged as the most likely causes of APOBEC3-mediated mutations in cancer3,5,22,23,31-35,37. Among APOBEC3 family members, expression levels of APOBEC3A and APOBEC3B correlate most strongly with mutational burdens in cancer genomes22,23,34,35. However, such correlations are generally weak, complicating assignment of the enzyme underlying the majority of the APOBEC3-associated mutations in cancer22,23,34,35. More prominent expression of APOBEC3B relative to APOBEC3A, strong deaminase activity in cell extracts and association between APOBEC3B expression and mutational burdens in cancer have been used to define APOBEC3B as a major mutator in breast and other cancers22,23. Despite these associations, cancers more frequently present with mutations enriched in extended sequence contexts otherwise preferred by APOBEC3A (YTCN contexts, where Y is a pyrimidine base) rather than contexts preferred by APOBEC3B (RTCN, in which R is purine)31. Such ‘APOBEC3A-associated cancers also harbor over ten-fold more mutations than ‘APOBEC3B-associated cancers, supporting the notion that APOBEC3A may be the more potent mutagen31. Refinement of the correlation analysis between APOBEC3 expression levels and change of conditions in deaminase activity assays, further indicates that APOBEC3A may be a more potent mutator35.

In a direct test of the mutagenic activities of endogenous APOBEC3A and APOBEC3B, individual APOBEC3A and APOBEC3B genes were deleted from human cancer cell lines which generate APOBEC3-associated signature over time2,38. Deletion of APOBEC3A severely diminished mutation acquisition, especially in YTCN contexts, demonstrating that endogenous APOBEC3A can mediate prevalently found cytosine mutations enriched at YTCN contexts in cancer2. Deletion of both APOBEC3A and APOBEC3B caused the most severe reduction in APOBEC3-associated mutations without completely eliminating them indicating that APOBEC3B, and perhaps an additional APOBEC enzyme, contributes smaller mutation burdens2. In cell lines where APOBEC3A contributed the majority of mutations, its relatively weak activity could be detected against model probes in extracts and against model RNA substrates2, in agreement with prior work35,37. However, APOBEC3B protein levels and deaminase activity were substantially elevated compared to APOBEC3A in these cell lines indicating that these assays may have limited utility as readouts of relative APOBEC3 mutagenic activities2.

The use of surrogate assays of APOBEC3-mediated mutagenesis, continues to nominate either APOBEC3A, or APOBEC3B as a more prominent mutator in different settings in cancer3,5,29,30. Expression-based readouts in cancer can be complicated by tumor infiltrating immune cells, which can have high levels of APOBEC3 expression39. Thus, bulk tumor mRNA sampling may not accurately reflect APOBEC3 expression in tumor cells3. APOBEC3-associated mutational signatures can be acquired episodically over time in cancer cell lines, and some human cancer cell lines with signatures of historic APOBEC3 mutagenesis exhibit no evidence of continued or episodic APOBEC3-associated mutation acquisition38. Therefore, expression of individual APOBEC3 enzymes detected at the time of cancer cell sampling may not accurately reflect origins of detected mutations. Furthermore, protein-based assays for APOBEC3 detection remain limited due to the lack of reagents, while more commonly measured transcript abundance may be a poor predictor of corresponding protein levels. These challenges likely underlie the weak correlations reported between expression of APOBEC3 enzymes and APOBEC3-associated mutations in cancer22,23,34,35.

In vitro cytosine deamination assays, in which a labeled ssDNA oligonucleotide is incubated with extracts from cell lines have been widely used to measure APOBEC3 activities22,35(Fig. 2). However, non-uniformity of experimental conditions confounds interpretation of these assays. Important parameters of these assays include RNA-mediated inhibition of deaminase activity35,40 and variability in sequence context preference of APOBEC3 deaminases upon different probes33. Distinct predictions regarding mutagenic activities of APOBEC3A or APOBEC3B may emerge upon altering these parameters across experimental conditions22,33,35. Recent work suggests that quantitative measures of RNA editing at hotspot hairpin loops targeted by APOBEC3A may serve as a marker of ongoing APOBEC3A deamination activity37. However, an APOBEC3A-induced mutation within a cancer genome reflects a combination of APOBEC3A-mediated deamination and subsequent errors introduced or enabled by DNA-repair and DNA-replication processes. Indeed, DNA repair processes can modulate the spectra and burdens of APOBEC3-associated mutations2,41,42. While both RNA editing and in vitro DNA deamination assays are important tools to study deamination activities of APOBEC3 enzymes, they do not measure the extent to which deamination events are converted into genomic mutations. Thus, these assays may not always reflect accurate readouts of APOBEC3 mutagenesis upon a genome and the extent to which they predict APOBEC3-mediated genomic mutagenesis has yet to be tested.

Analysis of cytosine mutations at extended motifs has gained traction as a readout of activities of the major candidate mutators, APOBEC3A and APOBEC3B (Fig. 2). These include enrichment of mutations at, respectively, APOBEC3A- and APOBEC3B-preferred YTCN and RTCN tetranucleotides31 and quantification of ‘hotspot’ cytosine mutations in sequences predicted to form hairpin loops, which otherwise make up a minor proportion of total APOBEC3-associated mutations in cancer genomes, to infer mutagenesis by APOBEC3A32,33. Extended sequence analysis of mutations accumulating in human cancer cell lines2 showed that endogenous APOBEC3 enzymes exhibit similar sequence preferences to APOBEC3A and APOBEC3B overexpressed in yeast31. However, preferences of many other APOBEC3 family members, which can otherwise cause mutations at TCN trinucleotides and thus possibly contribute signatures in cancer, have not been investigated at these sequence contexts and their activities cannot be excluded from these assays alone. Moreover, experiments in human cancer cell lines show that APOBEC3B can contribute smaller mutational burdens in the cell lines that display enrichment of mutations at YTCN-contexts mediated by APOBEC3A2. Therefore, extended sequence analyses cannot exclude minor contributions of some APOBEC3s. Furthermore, while hairpin loops represent preferred substrates for APOBEC3A and mutations at hairpin loops are less frequent in ‘APOBEC3B-associated’ cancers, APOBEC3B can be active upon hairpin loops31,33,43. Thus, the specificity of this assay for activity of APOBEC3A in human cancers requires further investigation43. Importantly, mutations at tetranucleotide sequence motifs and sequences predicted to form hairpin loops measure historically acquired APOBEC3-associated mutations and do not inform on whether mutagenesis remains active in samples under analysis. In addition, while these assays can point towards historic APOBEC3 activities in major clonal lineages captured in cancers under analysis, relative contributions of individual APOBEC3 enzymes may vary between individual cancer cell lineages and change during cancer evolution2.

Defined readouts of active mutagenesis that distinguish individual APOBEC3 enzymes are necessary to detect mutagenic activities of the relevant APOBEC3 deaminases in tumors and to enable experimental investigations into causes and functional consequences of APOBEC3 misregulation during cancer evolution. Human cancer cell lines with ongoing acquisition of APOBEC3-associated mutational signatures represent a suitable set of models to establish readouts that translate into active acquisition of the relevant mutations in cancer cells38. Deletion of individual APOBEC3 enzymes in such cell lines can be combined with overexpression of APOBEC3 enzymes across additional experimental models to further refine abilities of relevant assays to measure mutagenesis by individual APOBEC3 enzymes upon a genome2,13,32. It is possible that a combination of assays will be required to adequately identify active APOBEC3 mutagenesis. Once the optimal readouts of APOBEC3-mediated mutation acquisition have been determined in controlled experimental systems, the next challenge will be to assess their potential as clinical biomarkers of active mutagenesis by individual enzymes in human cancers.

Addressing impact of APOBEC3 enzymes on cancer evolution

Endogenous APOBEC3 enzymes can drive acquisition of APOBEC-associated signatures prevalent in cancer genomes2. Germline polymorphisms in the APOBEC3 locus are associated with increased mutational burdens and elevated cancer risk44-47. Expression of the human APOBEC3A transgene can promote tumorigenesis in mice predisposed to colon and liver cancers9,48,49. APOBEC3-associated mutations have been found in metastatic cancers and in subclonal branches of tumor phylogenies50-55. Driver mutations have been detected in APOBEC3-associated sequence contexts at different stages of cancer evolution, including in therapy resistance29,30,50-53,55-58. APOBEC3-associated signatures can continue to be acquired in human cancer cell lines over long periods of time38. These studies collectively provide support for the concept that APOBEC3 mutagenesis drives cancer evolution and contributes associated phenotypes, such as tumor heterogeneity, metastasis and resistance, and that its inhibition may confer therapeutic benefit (Fig 1B). However, therapeutic pursuit depends on establishing causal relationships between mutagenesis by endogenous APOBEC3 deaminases and cancer evolution, and on demonstrating that perturbing mutagenesis by individual enzymes can alleviate associated phenotypes, in preclinical models that closely recapitulate physiological settings where APOBEC3 inhibition is predicted to confer therapeutic benefit.

First, validation of associations between individual APOBEC3 enzymes and mutational signatures detected in various contexts, such as in different types of primary or therapy resistant cancers, requires experimental demonstration that nominated endogenous enzymes generate the relevant signatures in such contexts. Deletion of APOBEC3A from a panel of breast cancer and lymphoma cell lines diminished acquisition of the majority of APOBEC3-associated mutational signatures2. APOBEC3 enzymes can be systematically deleted across cell lines from additional cancer types and upon treatment with relevant therapies, such as standard of care or therapies associated with APOBEC3-mediated resistance5,29,30, to ascertain the relevant mutator enzymes across a broader spectrum of contexts where APOBEC3-associated signatures have been detected (Fig. 3). Second, once the relevant mutators have been experimentally validated, their impact on cancer evolution should be investigated across preclinical models that closely recapitulate settings where APOBEC3 mutagenesis has been linked to cancer evolution (Fig. 3). Critically, the hypothesis that APOBEC3 inhibition may confer therapeutic benefit and diminish the phenotypes associated with cancer cell evolution, such as sensitivity to therapy, metastatic potential and tumor heterogeneity, should be tested by assessing the relevant phenotypes upon perturbing mutagenesis by the relevant enzymes. Unlike humans, mice encode only a single APOBEC3 enzyme59. Therefore, mimicking human APOBEC3 mutagenesis in murine models relies on expression of the human APOBEC3 genes. Generation of transgenic models should be guided by human data. The choice of transgenic systems should be directed towards modeling mutagenesis in tissues where APOBEC3-associated mutational signatures have been observed in cancer. Reconstructions of the evolutionary trajectories of tumor and normal tissues60-62 will likely unveil genetic backgrounds upon which APOBEC3-associated mutations commonly occur in different tissues. Such backgrounds can be engineered into transgenic mouse models, to recapitulate the settings in which APOBEC3 mutagenesis is hypothesized to drive cancer evolution and associated phenotypes. APOBEC3 mutagenesis in mouse models should be recapitulated by ectopic expression of human APOBEC3 enzymes that were a priori established as responsible mutators in human cell models of the cancer type and/or therapy setting under analysis. Complementary studies using depletion of the relevant enzymes in patient-derived xenografts (PDX) and human cancer cell lines with active APOBEC3 mutagenesis are important to mitigate the potential neomorphic effects of APOBEC3 overexpression in mice. These experiments can be combined with treatments with relevant therapeutics to further mimic clinical settings in which APOBEC3 inhibition is speculated to confer therapeutic benefit.

Figure 3. Addressing impact of APOBEC3 enzymes on cancer evolution.

Figure 3.

APOBEC3 mutators can be identified by knockouts (KO) of individual enzymes in human cell lines, which actively acquire APOBEC3-associated mutations, from cancer types commonly presenting with APOBEC3-associated signatures (percentages of cancers presenting with APOBEC3-associated signatures in examplar cancer types are indicated). To detect the impact of APOBEC3 deletions on mutation acquisition, genomes of single-cell derived parent clones can be compared to genomes of their respective daughters obtained after defined in vitro periods. To investigate the roles of APOBEC3 mutagenesis in cancer cell evolution, identified APOBEC3 mutators can be expressed in murine models, which lack most human APOBEC3 gene orthologs. APOBEC3 expression can be performed upon genetic backgrounds and in tissues in which APOBEC3-associated mutations present in human cancers. Tumors that may result from APOBEC3 activities can be profiled for driver mutations and those can be compared to driver mutations in human cancers. To control for neomorphic effects, experiments in transgenic models can be accompanied by experiments in patient-derived xenografts and human cancer cell lines with active APOBEC3 mutagenesis. Analyses can be performed in cancers and models of various stages of the disease and/or upon therapy treatment to investigate the ability of APOBEC3 mutagenesis to contribute to different stages of cancer cell evolution, such as tumor heterogeneity, metastasis and resistance.

Third, therapeutic windows during which inhibition of APOBEC3 mutagenesis may confer the most effective clinical benefit should be investigated, by conducting experiments in preclinical models representing various stages of the disease. Additionally, the driver mutations arising in APOBEC3-associated sequence contexts in preclinical models should be compared to drivers identified in matching types of human cancers and therapy settings, to establish relevance of such mutations in human cancer cell evolution. APOBEC3 enzymes are likely sources of driver mutations occurring at TCN trinucleotides53. In classifying driver mutations as APOBEC3-induced in human cancers and experimental models, it will be relevant to statistically account for signatures of other mutational processes that can contribute mutations at TCN contexts58,63,64, such as signatures associated with ultraviolet light exposure and mutations in polymerase ε, particularly in cancers where such processes contribute high mutational burdens. Furthermore, identification of APOBEC3-associated driver mutations should take into account sequence features such as the potential to form DNA hairpins, which are preferred substrates for APOBEC3A32,33 and thus recurrent mutations at such sequences may not reflect positive selection.

Additionally, APOBEC3A and APOBEC3B have been implicated in induction of chromosomal instability65,66. Expression of APOBEC3A in avian cells was recently shown to induce genomic deletions in addition to SBS signatures13. Therefore, putative non-SBS alterations should be taken into account when investigating APOBEC3-mediated driver mutations. In addition, APOBEC3 activities may contribute to tumor progression by other means. For example, APOBEC3B activity in estrogen receptor (ER)-positive breast cancer cell lines can be directed to ER binding regions and mediate chromatin remodeling that promotes expression of ER target genes67, providing a possible mechanistic basis for association between APOBEC3B expression and poor survival of ER+ breast cancer patients19,67,68.

APOBEC3 enzymes have been experimentally linked to acquired resistance to tyrosine kinase inhibitors in lung cancer29,30. Such efforts should be expanded across a broader range of cancer types and therapy settings to investigate existing predictions about the impact of APOBEC3 mutagenesis on cancer cell evolution phenotypes, which await experimental validation16-28. Overall, investigations across models that closely recapitulate physiological settings in which APOBEC3 mutagenesis is active in cancer will be critical to assess the impact of individual APOBEC3 mutators on cancer evolution and associated phenotypes such as tumor heterogeneity, resistance and metastasis. Identification of the specific readouts of active APOBEC3 mutagenesis discussed in the previous section will accelerate such efforts.

Addressing (mis)regulation of APOBEC3 mutagenesis

The processes that regulate and misregulate APOBEC3 mutagenesis are key knowledge gaps that present barriers to therapeutic opportunities. Available data suggests that APOBEC3 mutagenesis may be instigated by both endogenous and environmental factors. APOBEC3-associated mutational signatures accumulate in some human cancer cell lines in the absence of obvious environmental stressors, indicating that APOBEC3 mutagenesis can arise from endogenous misregulation in cancer cells38. APOBEC3-associated signatures can be acquired episodically, rather than continuously, over time in individual cancer cell line lineages, suggesting that endogenous instigators of mutagenic APOBEC3 behavior may be intermittent38. Episodic APOBEC3 misregulation may account for highly variable APOBEC3-associated mutational burdens among phylogenetic branches of primary tumors54,69 and healthy bronchial cells70. On the other hand, multiple environmental factors, such as viral infections, genotoxic stress, some targeted therapies and cytotoxic drugs, can induce APOBEC3 expression and/or deamination activities in experimental models5,27-30,47,71-73. Although inductions of APOBEC3 expression and/or activity may not always translate into active genomic mutagenesis, some of the exogenous stressors, such as tobacco smoking, cytotoxic agents and some targeted therapies, have been associated with increased APOBEC3 mutational burdens28-30,74,75. Furthermore, APOBEC3-associated signatures are prevalent in cancers associated with human papillomavirus (HPV) otherwise restricted by APOBEC3 enzymes, as well as enriched in HPV-positive, compared to HPV-negative, head and neck cancers1,76. Thus, APOBEC3-associated signatures in cancer genomes may in part represent collateral damage from a response originally directed towards virus restriction76. However, APOBEC3-associated signatures have been detected in HPV-negative cancers and in many cancer types where viral infections are not apparent, while they have not been commonly observed in cancer types associated with viruses that can be restricted by APOBEC3 enzymes, such as in hepatitis-associated hepatocellular carcinoma1,56,76. Therefore, factors other than viral infections likely contribute APOBEC3 misregulation in many cancers and viral infections may not always instigate APOBEC3 misregulation. In conclusion, environmental factors likely trigger APOBEC3 mutagenesis, but understanding the relative contributions of environmental and endogenous factors to APOBEC3 mutagenesis will require further investigation.

The molecular details of how APOBEC3 dysfunction translates into genomic mutations are largely unknown, but likely include a combination of APOBEC3 expression, subcellular localization, availability of the genomic substrate, interacting proteins and DNA repair. The mechanisms may vary depending on the exogenous or endogenous instigator of aberrant APOBEC3 behavior. Expression of APOBEC3 genes in malignant and healthy tissues varies among family members34,77-79. While many prior studies have evaluated factors that activate the innate immune function of APOBEC3 enzymes, the signaling pathways that control physiological APOBEC3 expression are not completely understood39. Furthermore, multiple signaling pathways, such as interferon and PKC/NF-kB signaling, can induce expression of major mutator candidates APOBEC3A, APOBEC3B or both, in normal and malignant cells depending on cell type or context5,27,29,30,39,71. It is possible that contexts common in cancer cells promote misregulated APOBEC3 expression. For example, APOBEC3 induction in cancer could result from activation of DNA damage signaling pathways in response to replication stress, DNA breaks, or chemotherapeutic agents28,74. Further, the APOBEC3 genes may be regulated by p53 in response to DNA damage. While some APOBEC3 enzymes may be responsive to p53 activation, others such as APOBEC3B appear to be repressed by p53 and may therefore be misregulated by tumor-associated p53 mutants80-82. Overall, the signaling pathways that elevate APOBEC3 expression in cancer remain incompletely understood and it is not clear to what extent APOBEC3 upregulation translates into mutagenesis upon a cancer genome. Furthermore, post-transcriptional and post-translational modifications of APOBEC3 transcripts and proteins may play contributing roles, but have not been explored thoroughly.

APOBEC3 activity on the cellular genome may further be regulated through access to genomic ssDNA, for example, via cellular localization of APOBEC3 enzymes. Some family members are exclusively nuclear (e.g. APOBEC3B) and some are predominantly cytoplasmic (e.g. APOBEC3G), while others are found in both nuclear and cytoplasmic compartments (e.g. APOBEC3A)83,84. The episodic nature of APOBEC3 mutagenesis38 calls for further investigation into the possibility that aberrant localization of these enzymes occurs transiently, which may not be captured by standard assays. Further, availability of ssDNA substrate may represent a relevant barrier or facilitator for mutagenesis. Deamination substrates may be impacted by factors such as replication fork progression, DNA resection, break-induced replication, and chromatin conformation8,28,41,74,85-88. For example, associations from human cancer data and experiments in model systems implicate replication stress in increased APOBEC3 mutagenesis28,74,89. ssDNA generated during telomere crisis, a period of genomic instability that occurs following depletion of the telomere reserve, enables APOBEC3B-mediated generation of kataegis90. In this system APOBEC3B-dependent mutagenesis occurred independently of increased APOBEC3B mRNA or protein levels. Instead, APOBEC3B was shown to target available ssDNA that accumulated in aberrant nuclear compartments that formed as a result of chromosomal instability during telomere crisis. Additionally, ssDNA binding proteins such as RPA or RNA polymerase may limit APOBEC3 access to genomic substrates91.

APOBEC3 activities may also be regulated via protein interactions. Interaction between APOBEC3A and the cellular CCT chaperonin complex was recently found to minimize deaminase-induced DNA damage and cytotoxicity92. Viral oncoproteins provide further insights into how APOBEC3 activities may be regulated via protein interactions, for example via redirecting ubiquitination93 or cellular localization94. Finally, the DNA repair and replication processes involved in processing of APOBEC3-induced DNA edits can modulate the final burden and types of APOBEC3-associated mutations incurred on a cancer genome2,41,42. For example, deletion of the translesion polymerase REV1 diminishes large amounts of APOBEC3-associated signatures, while deletion of the uracil DNA glycosylase (UNG) decreases APOBEC3-associated transversion mutations2.

Overall, combinations of endogenous and environmental processes at different molecular levels likely determine the dynamics of regulation and misregulation of APOBEC3 mutagenesis upon a genome. Molecular, proteomic, and epidemiologic characterization is required to define the processes that ultimately regulate and misregulate APOBEC3-associated mutational burdens observed in cancer genomes. Improving the readouts and models of APOBEC3 mutagenesis discussed above will facilitate these efforts. Initially, the instigators of the APOBEC3 misregulation and molecular levels at which misregulation translates into mutagenesis of the human genome must be defined. Subsequent characterization of the factors involved in APOBEC3 misregulation at the relevant molecular levels may identify additional targets to halt APOBEC3-mediated mutation acquisition in cancer. Understanding the ways in which misregulation in cancer may differ from regulatory mechanisms in healthy tissues will help prioritize strategies to limit adverse effects.

Addressing physiological APOBEC3 functions

The APOBEC3 gene locus has undergone a rapid evolutionary expansion resulting in seven genes in primates59. The expansion is thought to be underlined by the selective pressure on the APOBEC3 genes from their retroviral and retrotransposon targets95-97. An evolutionary advantage would predict deleterious effects upon APOBEC3 inhibition. However, current knowledge of the physiological roles of APOBEC3 enzymes across human tissues is incomplete, limiting predictions regarding consequences of targeting APOBEC3 deaminase activities.

The best-defined function of the APOBEC3 family is in innate-immune restriction of retroviruses and retroelements. The first APOBEC3 enzyme identified, APOBEC3G, was characterized by its ability to restrict Vif-deficient human immunodeficiency virus (HIV) through deamination of the viral genome in its cDNA intermediate phase98,99. Activities of APOBEC3G and other APOBEC3 members have now been linked to additional targets, including ssDNA viruses, double-stranded (ds) DNA viruses, and RNA viruses1,100. The major candidate mutators in cancer, APOBEC3A and APOBEC3B, have been reported as viral restriction factors against a narrow range of pathogens101. Despite recent progress, in vivo data regarding APOBEC3-mediated viral restriction are lacking. Furthermore, the functions of APOBEC3 enzymes beyond the immune system have not been thoroughly investigated.

Limited available evidence suggests that APOBEC3 inhibition may not severely compromise overall health. APOBEC3 knockout mice exhibit increased susceptibility to mouse retrovirus infection, but normal viability102,103. Furthermore, the germline APOBEC3B polymorphism, which effectively deletes APOBEC3B and fuses APOBEC3A to the 3’UTR of APOBEC3B, is common in some ancestries44. It is thus possible that there is redundancy among cellular pathways that affect virus and retroelement restriction. However, the APOBEC3B deletion polymorphism has been associated with increased mutational burdens and susceptibility to some cancer types within specific ancestries45,47,104-107. The increased mutational burdens in carriers of the APOBEC3B polymorphism may result from production of a stabilized version of the APOBEC3A transcript108. APOBEC3B loss can increase APOBEC3A protein levels, activity, and APOBEC3A-associated mutational burdens in some cancer cell lines that do not form the APOBEC3A-APOBEC3B 3'UTR hybrid transcript2. These results raise a counterintuitive possibility that the absence of some APOBEC3 members may result in higher overall mutational burdens, further highlighting the need to investigate interactions between APOBEC3 enzymes.

More thorough examination of physiological APOBEC3 functions will determine the consequences of perturbing APOBEC3 activities. Future investigations should include population-based studies of the risk-factors associated with APOBEC3 polymorphisms and systematic assessments of phenotypic and genotypic consequences of APOBEC3 perturbation in healthy and malignant tissues of model organisms and human cell systems.

Conclusion

Therapeutic approaches based on inhibiting APOBEC3 activities in cancer are commonly discussed, and inhibitor development is underway16-28. However, critical aspects of APOBEC3 biology remain unknown or rely on associations which have not been validated in preclinical models that closely resemble human cancer physiology. This lack of understanding thus limits predictions pertaining to efficacy and safety of proposed inhibitor approaches. We have highlighted these gaps and outlined experimental strategies to address them. Ultimately, effective investigations into the potential to inhibit APOBEC3 mutagenesis as a therapeutic strategy in cancer depend on re-prioritization of experimental approaches and experimental validation of associations made between APOBEC3 activities and cancer evolution. Additional proposals aimed at therapeutic exploitation of APOBEC3 activities in cancer have emerged. Those include induction of APOBEC3 mutagenesis to increase the efficacy of immune checkpoint blockade17,51,109,110 and exploiting synthetic lethality opportunities arising from APOBEC3-mediated DNA damage111-114. Future studies into APOBEC3 biology will facilitate understanding the potential of these alternative therapeutic avenues.

Acknowledgements

M.P.’s research was supported by the European Molecular Biology Organization Long-Term Fellowship (ALTF 760–2019) and is supported by the National Cancer Institute (NCI) (CA270102). J.M.’s laboratory is supported by the NCI (CA212290, CA008748, CA261183, CA270102), the Pew Charitable Trusts, the V Foundation, the Starr Cancer Consortium, the Emerald Foundation and the Geoffrey Beene and Ludwig Centers at the MSKCC. A.M.G.’s lab is supported by the NCI (K08CA21299), the Department of Defense (CA2000867), the American Cancer Society, the Cancer Research Foundation, and the Children’s Discovery Institute. Research on APOBEC enzymes in the lab of M.D.W. was supported by grants from the National Institutes of Health (CA181259 and CA185799). We thank Mary O’Reilly and Elena Berg from the Broad Institute’s Pattern team for figure graphics.

Footnotes

Competing interests

M.P. is a shareholder in Vertex Pharmaceuticals and a compensated consultant through GLG Network. J.M. has received consulting fees from Ono Pharmaceutical Co. J.M.'s spouse is an employee of and has equity in Bristol Myers Squibb. M.P and J.M. are inventors of the patent application entitled “Tracking APOBEC mutational signatures in tumor cells” (patent pending). The remaining authors declare no competing interests.

Data availability statement

No new data were generated for the manuscript.

References

  • 1.Harris RS & Dudley JP APOBECs and virus restriction. Virology 479-480, 131–145 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Petljak M et al. Mechanisms of APOBEC3 mutagenesis in human cancer cells. Nature (2022) doi: 10.1038/s41586-022-04972-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Petljak M & Maciejowski J Molecular origins of APOBEC-associated mutations in cancer. DNA Repair 94, 102905 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Green AM & Weitzman MD The spectrum of APOBEC3 activity: From anti-viral agents to anti-cancer opportunities. DNA Repair 83, 102700 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Granadillo Rodríguez M, Flath B & Chelico L The interesting relationship between APOBEC3 deoxycytidine deaminases and cancer: a long road ahead. Open Biol. 10, 200188 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mas-Ponte D & Supek F DNA mismatch repair promotes APOBEC3-mediated diffuse hypermutation in human cancers. Nat. Genet 52, 958–968 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bergstrom EN et al. Mapping clustered mutations in cancer reveals APOBEC3 mutagenesis of ecDNA. Nature 602, 510–517 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Roberts SA et al. Clustered mutations in yeast and in human cancers can arise from damaged long single-strand DNA regions. Mol. Cell 46, 424–435 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Alexandrov LB et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nik-Zainal S et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Degasperi A et al. Substitution mutational signatures in whole-genome–sequenced cancers in the UK population. Science vol. 376 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.DeWeerd RA et al. Prospectively defined patterns of APOBEC3A mutagenesis are prevalent in human cancers. Cell Rep. 38, 110555 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Harris RS, Petersen-Mahrt SK & Neuberger MS RNA editing enzyme APOBEC1 and some of its homologs can act as DNA mutators. Mol. Cell 10, 1247–1253 (2002). [DOI] [PubMed] [Google Scholar]
  • 15.Hultquist JF et al. Human and rhesus APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H demonstrate a conserved capacity to restrict Vif-deficient HIV-1. J. Virol 85, 11220–11234 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Olson ME, Harris RS & Harki DA APOBEC Enzymes as Targets for Virus and Cancer Therapy. Cell Chemical Biology vol. 25 36–49 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Venkatesan S et al. Perspective: APOBEC mutagenesis in drug resistance and immune escape in HIV and cancer evolution. Ann. Oncol 29, 563–572 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Swanton C, McGranahan N, Starrett GJ & Harris RS APOBEC Enzymes: Mutagenic Fuel for Cancer Evolution and Heterogeneity. Cancer Discov. 5, 704–712 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Law EK et al. The DNA cytosine deaminase APOBEC3B promotes tamoxifen resistance in ER-positive breast cancer. Sci Adv 2, e1601737 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kvach MV et al. Inhibiting APOBEC3 Activity with Single-Stranded DNA Containing 2’-Deoxyzebularine Analogues. Biochemistry 58, 391–400 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Barzak FM et al. Selective inhibition of APOBEC3 enzymes by single-stranded DNAs containing 2’-deoxyzebularine. Org. Biomol. Chem 17, 9435–9441 (2019). [DOI] [PubMed] [Google Scholar]
  • 22.Burns MB et al. APOBEC3B is an enzymatic source of mutation in breast cancer. Nature 494, 366–370 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Burns MB, Temiz NA & Harris RS Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat. Genet 45, 977–983 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Harris RS Molecular mechanism and clinical impact of APOBEC3B-catalyzed mutagenesis in breast cancer. Breast Cancer Res. 17, 8 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kuong KJ & Loeb LA APOBEC3B mutagenesis in cancer. Nature Genetics vol. 45 964–965 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Olson ME et al. Oxidative Reactivities of 2-Furylquinolines: Ubiquitous Scaffolds in Common High-Throughput Screening Libraries. J. Med. Chem 58, 7419–7430 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Leonard B et al. The PKC/NF-κB signaling pathway induces APOBEC3B expression in multiple human cancers. Cancer Res. 75, 4538–4547 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kanu N et al. DNA replication stress mediates APOBEC3 family mutagenesis in breast cancer. Genome Biol. 17, 185 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Isozaki H et al. APOBEC3A drives acquired resistance to targeted therapies in non-small cell lung cancer. bioRxiv (2021) doi: 10.1101/2021.01.20.426852. [DOI] [Google Scholar]
  • 30.Caswell DR et al. The role of APOBEC3B in lung tumour evolution and targeted therapy resistance. bioRxiv 2020.12.18.423280 (2022) doi: 10.1101/2020.12.18.423280. [DOI] [Google Scholar]
  • 31.Chan K et al. An APOBEC3A hypermutation signature is distinguishable from the signature of background mutagenesis by APOBEC3B in human cancers. Nat. Genet 47, 1067–1072 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Langenbucher A et al. An extended APOBEC3A mutation signature in cancer. Nat. Commun 12, 1602 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Buisson R et al. Passenger hotspot mutations in cancer driven by APOBEC3A and mesoscale genomic features. Science 364, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Roberts SA et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat. Genet 45, 970–976 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cortez LM et al. APOBEC3A is a prominent cytidine deaminase in breast cancer. PLoS Genet. 15, e1008545 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Starrett GJ et al. The DNA cytosine deaminase APOBEC3H haplotype I likely contributes to breast and lung cancer mutagenesis. Nat. Commun 7, 1–13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Jalili P et al. Quantification of ongoing APOBEC3A activity in tumor cells by monitoring RNA editing at hotspots. Nat. Commun 11, 2971 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Petljak M et al. Characterizing Mutational Signatures in Human Cancer Cell Lines Reveals Episodic APOBEC Mutagenesis. Cell 176, 1282–1294.e20 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Covino DA, Gauzzi MC & Fantuzzi L Understanding the regulation of APOBEC3 expression: Current evidence and much to learn. J. Leukoc. Biol 103, 433–444 (2018). [DOI] [PubMed] [Google Scholar]
  • 40.Xiao X et al. Structural determinants of APOBEC3B non-catalytic domain for molecular assembly and catalytic regulation. Nucleic Acids Res. 45, 7540 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Taylor BJ et al. DNA deaminases induce break-associated mutation showers with implication of APOBEC3B and 3A in breast cancer kataegis. Elife 2, e00534 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Volkova NV et al. Mutational signatures are jointly shaped by DNA damage and repair. Nat. Commun 11, 2169 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jarvis MC et al. Mutational impact of APOBEC3B and APOBEC3A in a human cell line. doi: 10.1101/2022.04.26.489523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kidd JM, Newman TL, Tuzun E, Kaul R & Eichler EE Population stratification of a common APOBEC gene deletion polymorphism. PLoS Genet. 3, e63 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nik-Zainal S et al. Association of a germline copy number polymorphism of APOBEC3A and APOBEC3B with burden of putative APOBEC-dependent mutations in breast cancer. Nat. Genet 46, 487–491 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Long J et al. A common deletion in the APOBEC3 genes and breast cancer risk. J. Natl. Cancer Inst 105, 573–579 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Middlebrooks CD et al. Association of germline variants in the APOBEC3 region with cancer risk and enrichment with APOBEC-signature mutations in tumors. Nat. Genet 48, 1330–1338 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Law EK et al. APOBEC3A catalyzes mutation and drives carcinogenesis in vivo. J. Exp. Med 217, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Letouzé E et al. Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat. Commun 8, 1315 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kingston B et al. Genomic profile of advanced breast cancer in circulating tumour DNA. Nat. Commun 12, 2423 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Barroso-Sousa R et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann. Oncol 31, 387–394 (2020). [DOI] [PubMed] [Google Scholar]
  • 52.de Bruin EC et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346, 251–256 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.McGranahan N et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med 7, 283ra54 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jamal-Hanjani M et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. N. Engl. J. Med 376, 2109–2121 (2017). [DOI] [PubMed] [Google Scholar]
  • 55.Roper N et al. APOBEC Mutagenesis and Copy-Number Alterations Are Drivers of Proteogenomic Tumor Evolution and Heterogeneity in Metastatic Thoracic Tumors. Cell Rep. 26, 2651–2666.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Henderson S, Chakravarthy A, Su X, Boshoff C & Fenton TR APOBEC-mediated cytosine deamination links PIK3CA helical domain mutations to human papillomavirus-driven tumor development. Cell Rep. 7, 1833–1841 (2014). [DOI] [PubMed] [Google Scholar]
  • 57.Glaser AP et al. APOBEC-mediated mutagenesis in urothelial carcinoma is associated with improved survival, mutations in DNA damage response genes, and immune response. Oncotarget 9, 4537–4548 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cannataro VL, Mandell JD & Townsend JP Attribution of Cancer Origins to Endogenous, Exogenous, and Preventable Mutational Processes. Mol. Biol. Evol 39, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Conticello SG The AID/APOBEC family of nucleic acid mutators. Genome Biol. 9, 229 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Gerstung M et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Moore L et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020). [DOI] [PubMed] [Google Scholar]
  • 62.Li R et al. A body map of somatic mutagenesis in morphologically normal human tissues. doi: 10.1101/2020.11.30.403436. [DOI] [PubMed] [Google Scholar]
  • 63.Morganella S et al. The topography of mutational processes in breast cancer genomes. Nat. Commun 7, 11383 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vöhringer H, Van Hoeck A, Cuppen E & Gerstung M Learning mutational signatures and their multidimensional genomic properties with TensorSignatures. Nat. Commun 12, 3628 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Venkatesan S et al. Induction of APOBEC3 Exacerbates DNA Replication Stress and Chromosomal Instability in Early Breast and Lung Cancer Evolution. Cancer Discov. 11, 2456–2473 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wörmann SM et al. APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis. Nat Cancer 2, 1338–1356 (2021). [DOI] [PubMed] [Google Scholar]
  • 67.Periyasamy M et al. APOBEC3B-Mediated Cytidine Deamination Is Required for Estrogen Receptor Action in Breast Cancer. Cell Rep. 13, 108–121 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Sieuwerts AM et al. Elevated APOBEC3B correlates with poor outcomes for estrogen-receptor-positive breast cancers. Horm. Cancer 5, 405–413 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lee J-K et al. Clonal History and Genetic Predictors of Transformation Into Small-Cell Carcinomas From Lung Adenocarcinomas. J. Clin. Oncol 35, 3065–3074 (2017). [DOI] [PubMed] [Google Scholar]
  • 70.Yoshida K et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature 578, 266–272 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Oh S et al. Genotoxic stress and viral infection induce transient expression of APOBEC3A and pro-inflammatory genes through two distinct pathways. Nat. Commun 12, 1–17 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Warren CJ, Westrich JA, Van Doorslaer K & Pyeon D Roles of APOBEC3A and APOBEC3B in Human Papillomavirus Infection and Disease Progression. Viruses 9, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Starrett GJ et al. Polyomavirus T Antigen Induces APOBEC3B Expression Using an LXCXE-Dependent and TP53-Independent Mechanism. mBio vol. 10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hoopes JI et al. APOBEC3A and APOBEC3B Preferentially Deaminate the Lagging Strand Template during DNA Replication. Cell Rep. 14, 1273–1282 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alexandrov LB et al. Mutational signatures associated with tobacco smoking in human cancer. Science 354, 618–622 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Alexandrov LB et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Koning FA et al. Defining APOBEC3 expression patterns in human tissues and hematopoietic cell subsets. J. Virol 83, 9474–9485 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Refsland EW et al. Quantitative profiling of the full APOBEC3 mRNA repertoire in lymphocytes and tissues: implications for HIV-1 restriction. Nucleic Acids Res. 38, 4274–4284 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ng JCF, Quist J, Grigoriadis A, Malim MH & Fraternali F Pan-cancer transcriptomic analysis dissects immune and proliferative functions of APOBEC3 cytidine deaminases. Nucleic Acids Res. 47, 1178–1194 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Periyasamy M et al. p53 controls expression of the DNA deaminase APOBEC3B to limit its potential mutagenic activity in cancer cells. Nucleic Acids Res. 45, 11056–11069 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Menendez D, Nguyen T-A, Snipe J & Resnick MA The Cytidine Deaminase APOBEC3 Family Is Subject to Transcriptional Regulation by p53. Mol. Cancer Res 15, 735–743 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Periyasamy M et al. Induction of APOBEC3B expression by chemotherapy drugs is mediated by DNA-PK-directed activation of NF-κB. Oncogene 40, 1077–1090 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lackey L et al. APOBEC3B and AID have similar nuclear import mechanisms. J. Mol. Biol 419, 301–314 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lackey L, Law EK, Brown WL & Harris RS Subcellular localization of the APOBEC3 proteins during mitosis and implications for genomic DNA deamination. Cell Cycle 12, 762–772 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Haradhvala NJ et al. Mutational Strand Asymmetries in Cancer Genomes Reveal Mechanisms of DNA Damage and Repair. Cell 164, 538–549 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Seplyarskiy VB et al. APOBEC-induced mutations in human cancers are strongly enriched on the lagging DNA strand during replication. Genome Res. 26, 174–182 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Sakofsky CJ et al. Break-induced replication is a source of mutation clusters underlying kataegis. Cell Rep. 7, 1640–1648 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chan K & Gordenin DA Clusters of Multiple Mutations: Incidence and Molecular Mechanisms. Annu. Rev. Genet 49, 243–267 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Green AM et al. APOBEC3A damages the cellular genome during DNA replication. Cell Cycle 15, 998–1008 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Maciejowski J et al. APOBEC3-dependent kataegis and TREX1-driven chromothripsis during telomere crisis. Nat. Genet (2020) doi: 10.1038/s41588-020-0667-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Brown AL et al. Single-stranded DNA binding proteins influence APOBEC3A substrate preference. Sci. Rep 11, 21008 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Green AM et al. Interaction with the CCT chaperonin complex limits APOBEC3A cytidine deaminase cytotoxicity. EMBO Rep. e52145 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Malim MH & Bieniasz PD HIV Restriction Factors and Mechanisms of Evasion. Cold Spring Harb. Perspect. Med 2, a006940 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cheng AZ et al. Epstein-Barr virus BORF2 inhibits cellular APOBEC3B to preserve viral genome integrity. Nat Microbiol 4, 78–88 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.McLaughlin RN Jr, Gable JT, Wittkopp CJ, Emerman M & Malik HS Conservation and Innovation of APOBEC3A Restriction Functions during Primate Evolution. Mol. Biol. Evol 33, 1889–1901 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sawyer SL, Emerman M & Malik HS Ancient adaptive evolution of the primate antiviral DNA-editing enzyme APOBEC3G. PLoS Biol. 2, E275 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zhang J & Webb DM Rapid evolution of primate antiviral enzyme APOBEC3G. Hum. Mol. Genet 13, 1785–1791 (2004). [DOI] [PubMed] [Google Scholar]
  • 98.Sheehy AM, Gaddis NC, Choi JD & Malim MH Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature 418, 646–650 (2002). [DOI] [PubMed] [Google Scholar]
  • 99.Harris RS et al. DNA deamination mediates innate immunity to retroviral infection. Cell 113, 803–809 (2003). [DOI] [PubMed] [Google Scholar]
  • 100.Esnault C et al. APOBEC3G cytidine deaminase inhibits retrotransposition of endogenous retroviruses. Nature 433, 430–433 (2005). [DOI] [PubMed] [Google Scholar]
  • 101.Salter JD, Bennett RP & Smith HC The APOBEC Protein Family: United by Structure, Divergent in Function. Trends Biochem. Sci 41, 578–594 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Okeoma CM, Lovsin N, Peterlin BM & Ross SR APOBEC3 inhibits mouse mammary tumour virus replication in vivo. Nature 445, 927–930 (2007). [DOI] [PubMed] [Google Scholar]
  • 103.Mikl MC et al. Mice deficient in APOBEC2 and APOBEC3. Mol. Cell. Biol 25, 7270–7277 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Xuan D et al. APOBEC3 deletion polymorphism is associated with breast cancer risk among women of European ancestry. Carcinogenesis 34, 2240–2243 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Komatsu A, Nagasaki K, Fujimori M, Amano J & Miki Y Identification of novel deletion polymorphisms in breast cancer. Int. J. Oncol 33, 261–270 (2008). [PubMed] [Google Scholar]
  • 106.Gansmo LB et al. APOBEC3A/B deletion polymorphism and cancer risk. Carcinogenesis 39, 118–124 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Klonowska K et al. The 30 kb deletion in the APOBEC3 cluster decreases APOBEC3A and APOBEC3B expression and creates a transcriptionally active hybrid gene but does not associate with breast cancer in the European population. Oncotarget vol. 8 76357–76374 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Caval V, Suspène R, Shapira M, Vartanian J-P & Wain-Hobson S A prevalent cancer susceptibility APOBEC3A hybrid allele bearing APOBEC3B 3′UTR enhances chromosomal DNA damage. Nature Communications vol. 5 (2014). [DOI] [PubMed] [Google Scholar]
  • 109.Wang S, Jia M, He Z & Liu X-S APOBEC3B and APOBEC mutational signature as potential predictive markers for immunotherapy response in non-small cell lung cancer. Oncogene 37, 3924–3936 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Driscoll CB et al. APOBEC3B-mediated corruption of the tumor cell immunopeptidome induces heteroclitic neoepitopes for cancer immunotherapy. Nat. Commun 11, 790 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Green AM et al. Cytosine Deaminase APOBEC3A Sensitizes Leukemia Cells to Inhibition of the DNA Replication Checkpoint. Cancer Res. 77, 4579–4588 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Buisson R, Lawrence MS, Benes CH & Zou L APOBEC3A and APOBEC3B Activities Render Cancer Cells Susceptible to ATR Inhibition. Cancer Res. 77, 4567–4578 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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. 31, 107705 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Biayna J et al. Loss of the abasic site sensor HMCES is synthetic lethal with the activity of the APOBEC3A cytosine deaminase in cancer cells. PLoS Biol. 19, e3001176 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No new data were generated for the manuscript.

RESOURCES