Peer-Review Report by Anonymous https://med.jmirx.org/2025/1/e70039
Peer-Review Report by Anonymous https://med.jmirx.org/2025/1/e70041
Authors' Response to Peer-Review Reports https://med.jmirx.org/2025/1/e69307
Abstract
Background: The causes of breast cancer are poorly understood. A potential risk factor is Epstein-Barr virus (EBV), a lifelong infection nearly everyone acquires. EBV-transformed human mammary cells accelerate breast cancer when transplanted into immunosuppressed mice, but the virus can disappear as malignant cells reproduce. If this model applies to human breast cancers, then they should have genome damage characteristic of EBV infection.
Objective: This study tests the hypothesis that EBV infection predisposes one to breast cancer by causing permanent genome damage that compromises cancer safeguards.
Methods: Publicly available genome data from approximately 2100 breast cancers and 25 ovarian cancers were compared to cancers with proven associations to EBV, including 70 nasopharyngeal cancers, 90 Burkitt lymphomas, 88 diffuse large B-cell lymphomas, and 34 gastric cancers. Calculation algorithms to make these comparisons were developed.
Results: Chromosome breakpoints in breast and ovarian cancer clustered around breakpoints in EBV-associated cancers. Breakpoint distributions in breast and EBV-associated cancers on some chromosomes were not confidently distinguished (P>.05), but differed from controls unrelated to EBV infection. Viral breakpoint clusters occurred in high-risk, sporadic, and other breast cancer subgroups. Breakpoint clusters disrupted gene functions essential for cancer protection, which remain compromised even if EBV infection disappears. As CRISPR (clustered regularly interspaced short palindromic repeats)–like reminders of past infection during evolution, EBV genome fragments were found regularly interspaced between Piwi-interacting RNA (piRNA) genes on chromosome 6. Both breast and EBV-associated cancers had inactivated genes that guard piRNA defenses and the major histocompatibility complex (MHC) locus. Breast and EBV-associated cancer breakpoints and other variations converged around the highly polymorphic MHC. Not everyone develops cancer because MHC differences produce differing responses to EBV infection. Chromosome shattering and mutation hot spots in breast cancers preferentially occurred at incorporated viral sequences. On chromosome 17, breast cancer breakpoints that clustered around those in EBV-mediated cancers were linked to estrogen effects. Other breast cancer breaks affected sites where EBV inhibits JAK-STAT and SWI-SNF signaling pathways. A characteristic EBV-cancer gene deletion that shifts metabolism to favor tumors was also found in breast cancers. These changes push breast cancer into metastasis and then favor survival of metastatic cells.
Conclusions: EBV infection predisposes one to breast cancer and metastasis, even if the virus disappears. Identifying this pathogenic viral damage may improve screening, treatment, and prevention. Immunizing children against EBV may protect against breast, ovarian, other cancers, and potentially even chronic unexplained diseases.
doi:10.2196/50712
Keywords
Introduction
In the United States, over 40,000 women die from breast cancer each year [
, ]. The causes of the disease are not well understood, making prevention and treatment empirical and hazardous. At the time of breast cancer diagnosis, its causes are difficult to isolate from multiple risk factors. A human cancer virus is one such risk factor. A tumor virus does not cause cancer by itself [ ] but can make cancer more likely by inhibiting tumor suppressors [ ] or activating oncogenes. Viral damage then increases cancer risks via mutations and chromosome breaks. Epstein-Barr virus (EBV), also called human herpesvirus 4, infects at least 90% of humans as a lifelong infection, often acquired at an early age [ ], but the virus remains latent and asymptomatic in most people. EBV may be a risk factor for breast cancer. Active infection is significantly more prevalent in breast cancer tissues than in normal and benign controls [ ], increasing risk by 4.75- to 6.29-fold [ ]. EBV transformed human mammary epithelial cells in culture so that xenografts in immunosuppressed mice accelerated breast cancer. Once malignant transformation occurred, EBV was no longer required [ ], but the cells remain malignant.There has been no way to test the idea that EBV causes breast cancer and can then disappear. However, cancers in other tissues have proven relationships to EBV infection, so these known EBV-associated cancers can be compared to breast cancers at the genome level. Cancers with unambiguous EBV associations include nasopharyngeal cancer (NPC), EBV-positive diffuse large B-cell lymphoma (DLBCL), endemic Burkitt lymphoma (BL) [
], and gastric cancer (GC). Some genomic similarities between these EBV-associated cancers and breast cancer can be derived from the literature. In NPC, 100% of malignant cells are EBV positive [ ]. Over 64% of NPCs are deficient in a pathway that depends on the breast cancer susceptibility genes BRCA1 and BRCA2 [ ], which accurately repair DNA crosslinks and breaks via the homologous recombination pathway. This sprawling, interconnected pathway includes Fanconi anemia (FA) gene products and is often designated as the FA-BRCA pathway. In 126 patients with NPC, BRCA1 and BRCA2 were the most frequently mutated genes (55.5% and 33.3%, respectively) [ ]. NPC mutations interfere with innate immunity and constitutively activate an inflammatory response. Overexpressed nuclear factor–κB (NF-κB) is a hallmark of NPC, occurring in 90% of NPCs [ ]. Similarly, almost all stage-3 breast cancers overexpress NF-κB [ ].In NPC and the other known EBV-associated cancers, EBV inhibits the FA-BRCA pathway by various methods, including using viral microRNAs to downregulate BRCA1 [
], hijacking other pathway components [ , ], and destabilizing SMC5/6-mediated chromatin interactions [ , ]. In GC, EBV infection and FA-BRCA pathway status are mutually exclusive [ ], implying that EBV infection is approximately equivalent to disabling the FA-BRCA pathway. In DLBCL, the best prognostic marker is FA-BRCA pathway status [ ]. In DLBCL and endemic BL, EBV variant infection accompanies MYC translocations. These translocations drive the disease and make a characteristic replacement of normal MYC control elements with highly active immunoglobulin regulatory sequences [ , ]. MYC amplification is frequent in breast cancers that have inactive BRCA1 [ ].NPCs, DLBCLs, BLs, GCs, and breast cancers all have deficits in correctly repairing double-strand breaks and crosslinks. The compromised FA-BRCA pathway can produce chromosomes with too many centromeres. During cell division, mitotic spindles pull chromatids with multiple centromeres in too many directions, generating chromosome breaks to destabilize the human genome [
, ]. In breast cancer, these variations mark breakpoints at translocations and oncogene amplifications [ ].If EBV contributes to breast cancer, gene deficits in breast cancers and EBV-associated cancers should produce comparable changes in the human genome that do not depend on whether EBV infection persists. The aim of this study was to test for these virus-induced genome changes using bioinformatic calculations and analyses. The results could implicate EBV and its variants in disabling a variety of molecular and cellular safeguards that protect against breast cancer and its metastasis. Whether or not cancer develops in response to EBV infection depends on major histocompatibility complex (MHC) gene polymorphisms [
, ], so not everyone infected with EBV will develop cancer. In susceptible people, genome damage is permanent and does not require large numbers of viral particles, active infection, or continuing virus presence. Childhood immunization against selected EBV gene products may do much to prevent breast, ovarian, and other cancers.Methods
Datasets Used in the Analysis
Overview
The initial data for analysis came from literature searches for studies on breast and EBV-associated cancers with large numbers of participants, unrestricted access to genome information, and complete whole-genome analysis. The first criterion for including breast cancer data was published intrachain or interchain chromosome breakpoints from high-quality, peer-reviewed publications produced by world-class laboratories. The second criterion was the availability of sufficient DNA sequence data to specify the location of these chromosome breakpoints. The third criterion was that genome sequencing had been done on samples taken before treatment began. These publicly available DNA sequence data were chosen to encompass diverse genetics, subtypes, stages, grades, morphologies, and outcomes. Initially, breast cancers were separated only broadly into those with a likely hereditary component versus those without this component. The cancers had to include typical morphologies such as ductal carcinomas, lobular carcinomas, medullary carcinomas, and invasive carcinomas (ie, “no special type”). The included breast cancers were all primary stage-2 or stage-3 cancers. Although surgery usually removes these primary tumors, cells with only a few additional late mutations are responsible for seeding local recurrences or metastases, so primary and metastatic tumors are not very different [
]. Although the selected cancers are not a random sample representing all breast cancers [ ], they are likely to have chromosome instability originating from diverse typical causes.Specifically, the breast cancer data used came from 560 breast cancer genome sequences, familial cancer data from 78 patients, methylation data from 1538 breast cancers versus 244 controls, 243 triple-negative breast cancers, and 2658 human cancers [
- ]. Data also included 74 breast cancers from high-risk women who were typed as having BRCA1- or BRCA2-associated mutations or cancers diagnosed before the age of 40 years [ , ]. Another study of familial breast cancers contributed 65 familial breast cancers [ ]. Gene breakpoints for many interchromosomal and intrachromosomal translocations and breakpoints were obtained from the COSMIC (Catalog of Somatic Mutations in Cancer) website, as curated from original publications or original articles and their supplemental informationsupplemental information [ - ]. provides a glossary of the terms used in this paper.Breakpoints in Breast Cancers From High-Risk Women
Hereditary cancers were taken as breast cancers from women with a typed high-risk BRCA1 or BRCA2 mutation diagnosed before the age of 70 years. Cancers from patients with onset before the age of 50 years were also included to add more data, since these women are at high risk for an inherited, cancer-associated mutation. These patient samples were chosen based on descriptions in published data defining the breast cancer cohorts [
, ].Sporadic Breast Cancers
Sporadic breast cancers were taken as breast cancers diagnosed after the age of 70 years that did not have a known inherited mutation [
].Breast Cancer Subgroups
Human epidermal growth factor receptor 2 (HER2)–positive and triple-negative breast cancer data used for subgroup analysis were from original publications [
] and the COSMIC website.Exclusions
Male breast cancers were excluded.
Data Source for Ovarian Cancers
Data for breakpoints in ovarian cancers were downloaded from the COSMIC website. The cancers corresponded to “mixed adenosquamous ovarian carcinomas” and were arbitrarily taken from those with the largest number of structural variants. These cancers all had the prefix “AOCS-” with further identification numbers and BRCA mutation status in parentheses as follows: 170-1-8 (negative), 120-3-6 (BRCA2), 142-3-5 (negative), 139-1-5 (negative), 086-3-2 (negative), 147-1-1 (BRCA1 and BRCA2), 094‐6-X (BRCA1), 094-1-1 (BRCA1), 088-3-8 (negative), 139-6-3 (BRCA2), 150-3-1 (negative), 116-1-3 (negative), 155-3-5 (BRCA2), 093-3-6 (negative), 034-3-8 (BRCA1), 091-3-0 (BRCA1), 139-19-0 (BRCA2), 170-3-5 (negative), 114-1-8 (negative), 064-3-3 (negative), 064-1-6 (negative), 106-1-1 (BRCA1), 152‐1-X (BRCA1), and 134-1-5 (unknown).
Original Data Sources for Cancers With Known EBV Associations: NPCs, Lymphomas, and GCs
Overview
NPC chromosome breakpoint positions were retrieved from Bruce et al [
] for 70 primary tumors of the nasopharynx at stages 1-4C. The data came from whole-genome sequencing of “63 micro-dissected tumors, 5-patient derived xenografts, and two cell lines.” DLBCL breakpoints were collected from 88 patients with DLBCL (aged >60 y) [ ]. The MYC breakpoints included class I and II MYC translocation locus breakpoints defined in BL, encompassing areas far upstream of c-myc [ - ]. Downstream breakpoints included an enhancer region approximately 565 kilobases long on the nearest telomere side of the MYC coding sequence [ ]. Older data provided fusion sequences as Gencode Accession numbers [ ]. These fusion sequences were downloaded as FASTA files and copied to BLAST (Basic Local Alignment Search Tool) for placement on the human GRCh38/hg38 reference sequence. GCs with inferred EBV infection status came from 34 (20.2%) out of 168 samples subjected to whole-genome sequencing [ ].Selection Bias
As much as possible, selection bias was avoided by blindly selecting samples, replicating samples using cohorts from different publications, using the largest possible groups of samples, and avoiding convenience sampling. Some experiments used a newer dataset from 780 breast cancers [
] for comparisons to confirm that selection bias was unlikely.Recruitment
Data from genome sequence studies did not include specific recruitment procedures for patients with cancer. However, patients are typically recruited through hospitals and clinics with referrals from medical professionals. Patients provide informed consent to have their genomes sequenced and used for research and to integrate cancer genome sequence data into treatment decisions [
].Methods Used to Determine That DNA Breakpoints From Breast and Ovarian Cancers Clustered Around Breakpoints in EBV-Associated Cancers
Calculation of Distances Between Breakpoints in Breast and Ovarian Cancers Versus EBV-Related Cancers
Before combining or comparing datasets, they were all converted to the same genome version, usually GRCh38. The break position in breast cancer nearest to a break in NPC was taken as the Microsoft Excel XLOOKUP value for the number of base pairs (bp) from the closest NPC breakpoint 5’ to the breast cancer break or the NPC breakpoint 3’ to the breast cancer break, whichever was closer (
). For comparing a given breast cancer breakpoint A2 to EBV-associated cancer breakpoints B2 to B72, the initial algorithm to find the nearest 5’ break position was written in cell C2 as follows:=XLOOKUP($A2,$B$2:$B$72,$B$2:$B$72,0, −1,1). Changing −1 to +1 gave D2, the nearest 3’ position. Distance from the breast cancer breakpoint was then calculated as =MIN(ABS(C2-A2), ABS(D2-A2)). The same formulas were then continuously updated by Excel to calculate all other breast cancer comparisons in column A. Differences in the amount of data available for NPC versus breast cancer breakpoints complicated the calculations near chromosome telomeres. Several methods of handling these end regions made no discernible difference in the outcomes. For a 5000-bp window, an overflow window of 5,000,000 was used to limit the number of bins to a maximum of 1000. Another method of calculating distances between chromosome breakpoints in different cancers used the minimum of the absolute values of distances between breast cancers and the array of breakpoints in GCs, BLs, or NPCs. This method gave results identical to XLOOKUP values but was more convenient to compare clusters of breast cancer breakpoints to those in lymphoid and epithelial EBV-associated cancers. Hundreds of millions of calculations were repeated at least twice. Most of the calculations in this section are presented in .DNA Sequence Homology Analyses to Determine Breakpoints in Human Cancer Sequences That Resemble Viral Sequences
The NCBI BLASTn program (MegaBLAST) and database [
- ] were used to compare DNA sequence homologies around breakpoints in breast cancers to all available viral DNA sequences. E (“expect”) values are related to P values and represent the probability that a given homology bit score occurs by chance. E values <1×10–10 were considered significant homology. In many cases, E values were “0” (<1×10–180) and always far below 1×10–10. The virus DNA was retrieved from BLAST searches using “viruses (taxid:10239),” with human sequences, mouse sequences, and uncharacterized sample mixtures excluded. Different strains and isolates of the same virus were tested for human homology. Specifically, the HKHD40 and HKNPC60 variants were often considered together as “EBV.”Methods Used for Chromosome Comparisons of Breakpoints in Breast Cancers in High-Risk Women Versus Breakpoints in Sporadic Breast Cancer
The NCBI Genome Decoration page provided chromosome annotation software [
].Identifying Genes Around the Most Frequent EBV-Binding Site Locations and Tethering Sites
EBV nuclear antigen 1 (EBNA1)–binding location genome coordinates [
, ] were used to tabulate genes within or near anchoring sites where EBV docks on human DNA. Breaks in breast cancers were compared to the gene positions around their EBNA1-binding sites. The Palindrome Site Finder from NovoPro and the EMBOSS palindrome program were used to identify palindromic DNA sequences.Comparisons for Similarities Among Human Herpesviruses
EBV variants HKHD40 and HKNPC60 were compared to human herpesviruses in BLASTn by entering the terms “human gamma herpesvirus 4,” “herpesviridae,” and “herpesvirales.” Values with ≥2000 bp in common were selected. The EBV reference sequence was also tested against the following proven cancer viruses: human herpesvirus 8 (also called Kaposi sarcoma virus), herpes simplex virus 1, and human cytomegalovirus.
Locating Piwi-Interacting RNA Sequences as Evidence of Past EBV Infection
Piwi-interacting RNA (piRNA) locations were retrieved from the piRNA bank [
, ]. To compare the positions of piRNAs in virus homology versus genome position graphs, the midpoints of piRNA sequences were assigned arbitrary homology values. Positions of differentially methylated regions near breast cancer breakpoints on chromosome 6 [ ] were compared to breakpoint positions for 70 NPCs based on published data analyses [ ].Viral Sequences in Human Genomes as Hypermutation and Rearrangement Sites in Breast Cancers
A graph of viral sequences in humans against chromothripsis breaks in breast cancers was so complex that it resisted interpretation, so only the 5 viral sequences nearest the chromothripsis breaks were used. The viral sequences nearest high-confidence chromothripsis breaks were determined in 5 iterations as genome coordinates where XLOOKUP values gave the minimum distances. Distances between all virus homology start points were then compared to all chromothripsis breakpoints.
Methods of Data Analyses and Statistical Software
DNA flanking sequences at breakpoints were downloaded primarily from the GRCh38/hg38 version of the University of California, Santa Cruz Genome Browser as FASTA files and copied directly into BLAST. Results were checked against breakpoints in 101 triple-negative breast cancers from a population-based study [
]. The University of California, Santa Cruz Genome Browser’s Liftover function interconverted different versions of genome coordinates into GRCh38/hg38 coordinates.Statistics
Excel, SPSS (IBM Corp), StatsDirect, Visual Basic (Microsoft), and Python (Python Software Foundation) scripts were used for data analysis. Mann-Whitney U tests compared overall breakpoint distributions [
] and tested the hypothesis that breakpoint distributions were identical or at least roughly the same. The Mann-Whitney U test was chosen because the comparisons involved unequal numbers of breakpoints, and each observation was likely independent. P values >.05 were taken to indicate that identical distributions could not be excluded. Tests for normality included kurtosis and skewness values and evaluation by Shapiro-Francia and Shapiro-Wilk methods [ ] ( ). The Fisher exact test compared breakpoints in breast cancers to those in known viral cancers. The unpaired 2-tailed Student t test was used to compare the means of numbers of breast cancers with severe versus nil lymphocyte infiltrates, assuming the data approximated normality and that there were no extreme outliers. Both of these tests require independence and random sampling. All these test results are only approximate because they depend on underlying assumptions.Fragile Site Sequence Data
Positions of common fragile sites were retrieved from a database [
] and original publications [ ].Ethical Considerations
This study presents analyses of publicly available data without recruiting additional human or animal subjects. Because this study is a secondary analysis, it is exempt from institutional review board and ethics approval. The data are in the public domain and are available for independent research and analysis [
]. It is not necessary to obtain permission to reuse public data. The original informed consent allows secondary analysis without additional permission.Results
Breakpoints in Breast Cancers From High-Risk Backgrounds Clustered Around Breakpoints in NPC, an EBV-Mediated Cancer
EBV-mediated cancers such as NPC have defects in DNA repair and inflammatory pathways, resembling hereditary breast and ovarian cancers. To further characterize this resemblance, breakpoints in 70 NPC genomes were compared to breakpoints in 139 breast cancer genomes from high-risk women (BRCA1/BRCA2 mutation, familial concentration, or young age).
The distances from all breast cancer breakpoints to the nearest NPC breakpoints across the entire length of chromosome 1 produced results with so many points that they were difficult to interpret (
). Different laboratories collected these breakpoint data over many years. To allow for some variations, the data were grouped into 5000-bp increments (2×10–5 relative error). As shown in and , breast cancer breakpoints were most often clustered within 5000 bp of NPC breakpoints, but many breakpoints agreed much more closely. A total of 20 breast cancer breakpoints on chromosome 1 were within 500 bp of an NPC breakpoint, and several chromosomes had breast cancer and NPC breakpoints in essentially the same positions. As represented by Mann-Whitney U test results ( ), breast cancer and NPC breakpoint distributions were statistically the same (P>.05) for chromosomes 6, 7, 10, 13, 14, 15, 22, and X, but different on chromosome 1 and other chromosomes (P<.05).In contrast, liver cancer breakpoints at hepatitis B virus integration sites [
] differed from those in breast cancer or NPC ( ). No breaks in 114 liver cancers on chromosome 1 were within 5000 bp of breaks in any NPC; only one break on chromosome 6 in 61 liver cancers fit this window. According to a meta-analysis, the chance that breakpoints on chromosomes 1, 2, 6, and 8 were not within 5000 bp in liver cancer versus NPC was 4.4 (95% CI 1.9‐10). NPC and liver cancer did not have the same breakpoint distributions (P<.001).The above results revealed that breast cancer breakpoints in high-risk women were clustered near those in the EBV-associated cancer, NPC, on every chromosome. The next step was to decide whether these similarities depended on mutations in the breast cancer susceptibility genes, BRCA1 or BRCA2, by comparisons to sporadic breast cancers. The sporadic breast cancer group comprised 74 women, aged ≥70 years, with normal BRCA genes and no other known inherited, cancer-associated mutations [
]. Like breakpoints from high-risk women, many sporadic breast cancer breakpoints clustered around those in NPC ( ). Breakpoints in these sporadic breast cancers clustered at chromosomal locations similar to breast cancers from high-risk women, although the frequencies and distributions sometimes differed significantly. The patients with sporadic breast cancer were older than the high-risk women, arguing against age as responsible for similarity to NPC breakpoints.Viral Homologies Around Breakpoints in Mixed Adenosquamous Ovarian Carcinoma Also Clustered Around Breakpoints in EBV-Mediated Cancer
Ovarian cancer data enabled an additional test for EBV involvement in breast cancer because, like breast cancer, BRCA1 or BRCA2 mutations can also predispose patients to ovarian cancer [
]. Chromosome breakpoints in 25 mixed adenosquamous ovarian cancers were compared to breakpoints in NPCs. The results depicted in emulated breast cancer comparisons. Nearly half (12/25, 48%) the ovarian cancer cases had likely hereditary BRCA mutations. The remaining sporadic ovarian cancers gave the same results as the complete set but with less data. As in breast cancer, ovarian cancer breakpoint distributions clustered around NPC breakpoints, even without a hereditary BRCA1 or BRCA2 gene mutation driver.Breaks in Lymphomas Associated With EBV Infection Also Matched Breast Cancer and NPC
EBV drives lymphomas as well as NPCs. Based on epidemiologic research results, FA-BRCA pathways protect against lymphomas [
, ]. If EBV is genuinely associated with breast cancer breakpoints, then breakpoint positions in EBV-mediated lymphomas should also resemble those of breast and ovarian cancers. Because MYC gene rearrangements are characteristic of EBV-associated lymphomas, the first test of this idea was to survey virus-like sequences surrounding the MYC gene locus on the human reference genome. shows that MYC resides in a literal forest of retrovirus sequences (eg, human immunodeficiency virus type 1 [HIV1], feline leukemia virus, porcine endogenous retrovirus, and human endogenous retrovirus [HERV]) interspersed with EBV-like sequences.The concentration of virus sequences around MYC on chromosome 8 prompted the addition of the EBV-associated lymphoma DLBCL to breakpoint comparisons. As shown in
, the results revealed that hundreds of breast cancer and NPC breakpoints congregated around breakpoint positions in 88 DLBCLs [ ]. This agreement was consistent with other similarities between breast cancers and these EBV-associated cancers, including deficits in FA-BRCA pathway–mediated DNA repair by homologous recombination [ ] and NF-κB activation [ , - ].EBV is also a proven driver of at least one subset of BLs, typically those with MYC translocations. BL subsets can have mutations that impair homologous recombination [
], so results in revealed many breast cancer breakpoint positions near corresponding BL breakpoints. An older dataset from BLs [ ] had translocation breakpoints in the virus sequence–rich area near the MYC locus, agreeing with about 140 breast cancer breakpoints. Four different NPC breakpoints produced over 100 matches to BL translocation breakpoints, beginning at 8250 bp apart. An unpaired, 2-tailed t test did not support a statistically significant difference between BL and NPC breakpoints in this area (P=.69).Further tests were conducted to determine whether the functions of genes near clustered breakpoints supported a relationship between breast cancers and EBV-related cancers (GC [
], BL, and NPC). As illustrated in , breast cancer breakpoints on chromosomes 6, 8, 11, and 17 aggregated near positions where breakpoints occurred in EBV-associated cancers. Many aggregated breakpoints were in the same areas as genes that control inflammation, antiviral defenses, apoptosis, intermediate filaments, epigenetic and chromatin regulation, estrogen receptor activity, mitotic structures, and mitotic controls (Table S1 in ). Breast cancer breakpoints that clustered around EBV-associated cancer breakpoints were especially numerous on chromosome 17. One of these clusters marked in included the HER2 amplicon and the topoisomerase 2a gene, with BRCA1 and SMARCE1 genes nearby. SMARCE1 encodes a part of a chromatin regulation complex. Chromosome 17 breakpoints near CNTROB and CTC1 genes connect EBV to centriole and telomere malfunctions during mitosis (Table S1 in ). Rearrangements near breakpoints may cause over- or underexpression of nearby genes (Table S2 in ). Many additional correlations were also likely revealed in but were not investigated further.Results in this section show that breast cancer breakpoints clustered around breakpoints in additional EBV-associated cancers, where they affect critical functions needed to prevent breast cancer. Once these functions are compromised, cancer can occur without the continuing presence of EBV.
Genes at the Most Frequent EBV-Tethering Sites Clustered Around Breast Cancer Breakpoints
In preceding sections, breast and ovarian cancer breakpoints were found to distribute most frequently near characteristic sets of breakpoints associated with EBV-related cancers. The virus first attaches its EBNA1 protein to human DNA in the nucleus. Then, circular EBV episomes dock to this attached EBNA1 anchor. To test whether the initial EBNA1 attachment sites were related to breast cancer chromosome breakpoints, breast cancer breakpoints were compared to genes near EBV-docking sites. EBV-positive BL cells providing the data had up to 1569 EBV-docking sites on all chromosomes identified by 4C-chromatin capture experiments [
]. As shown in , the largest numbers of breast cancer breakpoints on most chromosomes clustered around the genes [ ] nearest to genes at EBV-docking sites. In support of these comparisons, graphical estimation of virus-tethering sites on chromosome 2 from chromatin capture data for these EBV-positive cells also agreed with breast cancer breakpoints ( ). In an unrelated study [ ], EBV-docking sites on chromosome 11 near known EBV anchor sites at the FAM-D and FAM-B genes were found near groups of breast cancer breakpoints, but imperfect palindrome sequences [ ] were more distant ( ). This finding independently supports the idea that EBV-docking sites are near breast cancer breakpoints. Results in this section raise the possibility that EBV directly contributes to breast cancer chromosome breakpoints and fragmentation.Breakpoints Occurred Near Human Sequences That Resemble Viruses in All Breast Cancers Tested
To further test whether EBV itself has some role in breaking chromosomes or altering their structures, human chromosomes were compared to all known viruses. As shown in
, the results showed that nearly every breast cancer likely had undergone breakages near EBV-like sequences. Chromosome 8 alone had 59,566 significant (>200) viral homology scores. Based on data from 128 patients with breast cancers and 43,491 unique breakpoints, breakpoints in 123 (96.1%) out of 128 breast cancers were within 10,000 bp of a virus sequence. In 106 patients, the virus was an EBV tumor variant (HKHD40 or HKNPC60) with 3086 matching human sequences. According to the Fisher exact test, chromosome 8 breakpoints and EBV variant sequence matches were not independent (P<.001).Many areas on other chromosomes also had 97% human-virus identity over nearly 2500 bp. It is implausible that this much similarity comes from EBV DNA being carried over into the human reference genome. Viral homology occurred with only a small, select portion of viral DNA [
]. Viral homologies were determined for a human genome in a homozygous karyotype, haploid cell line (46,XX) hydatidiform mole derived only from the paternal chromosomes in an X-bearing sperm cell after fertilization [ ]. Results still showed extensive homology between the mole and EBV variants HKHD40 and HKNPC60 ( ).HKHD40 and HKNPC60 variant sequences kept appearing in comparisons to human sequences, so these variants were tested against other herpesviruses to determine whether they were unusual. Hundreds of human gamma herpesvirus 4 variants were almost identical to HKHD40 and HKNPC60 over at least 2000 bp (
). The matching sets of viruses included many high-risk herpesvirus isolates from NPCs [ ]. Based on this information, HKHD40 and HKNPC60 strongly resembled other herpesvirus isolates, including many that confer high risks for NPC [ ]. These results show that humans have interacted extensively with EBV; the results are not due to EBV impurities in the human reference genome, and the human genome has had close relationships with oncogenic EBV forms.Evidence of Past EBV Infection
The evidence thus far supports a central hypothesis that EBV disables tumor suppressor mechanisms in breast cancer and can then disappear. This absence of viral particles is a significant experimental obstacle to testing this hypothesis. Unlike retroviruses, EBV and its variants do not have integrase enzymes, so EBV has no conventional way to insert itself into the human genome. EBV rarely integrates, with only one or two copies in BL cell lines [
].BLAST analysis found about 65,000 areas of strong homology (E<1×10–10) between the human reference genome and EBV. Because 65,000 is far more than realistic EBV integration events, it suggested the possibility that some EBV sequences were fragments created by a human version of the bacterial CRISPR (clustered regularly interspaced short palindromic repeats) system. As shown previously in
, breast cancers have breakpoints that cluster around breakpoints in EBV-associated cancers and involve MHC genes.MHC genes are encoded on chromosome 6p21.3 in a region that becomes a candidate for such a human CRISPR version. Variants of human leukocyte antigens (HLAs) in the MHC are strong risk factors for NPC infections [
] because HLAs are required to break down and display fragments of some antigens to the immune system. A total of 13 breast cancers listed on the COSMIC website had a deletion near this HLA region. About 23% of breast cancers had mutations directly affecting HLA class I or II genes. Many more breast cancers had indirect connections because they had damage to multiple genes that interact with HLAs or were otherwise essential for immunity. The MHC region also holds NFKB1L1, a negative regulator of the NPC overexpressed gene hallmark, NF-κB. The 139 breast cancers from high-risk women had 284 breakpoints at chromosome 6p21.3. Breakpoints in the 70 NPC cancers also clustered there, with 40 breakpoints within the 27,865,296-34,017,013 segment on chromosome 6. Variability in the inactivation of MHC genes reflects the extreme diversity of this region.In general, the bacterial CRISPR/Cas system loosely resembles the human piRNA system, so the distribution of piRNAs was graphed. As shown in
, hundreds of piRNA sequences cluster near the MHC region (at ~29.7‐33.3 megabases). The piRNA system is known to inactivate virus-derived transposons (related to HERVs) by methylating or cleaving them. The distribution of piRNA fragments was then compared to the distribution of viral DNA fragments in the MHC region of chromosome 6. reveals striking similarities in how remnants of exogenous and endogenous viruses distribute relative to piRNAs. Remnants of both virus types were homologous to the same human sequence, and both types were interspaced between piRNA sequences, sometimes right next to each other. Most of these sandwiches were at a regular interval or a multiple of a regular interval.This interspaced arrangement looked so much like CRISPR that it raised the question of whether piRNA defense mechanisms have inactivated some EBV variants in addition to their canonical role with endogenous viruses. Long stretches of endogenous transposon-like DNA sequences routinely matched exogenous viruses. As shown in
, the same human DNA interval had homology both to endogenous transposons (HERV) and exogenous viral sequences (EBV variants, stealth virus 1, chikungunya virus, BeAn 58058 virus, human papillomavirus [HPV] 16, HIV1, and HERV). This result shows that the piRNA system can store the same piece of DNA to protect DNA against these different viruses.Chromosome 6p21.3 also contains an EBV infection marker [
]. The marker was examined in 1538 breast cancers using existing methylation data [ ]. As indicated in , promoter methylation differed significantly from normal controls in the segment shown (30,523,984‐33,216,811 on chromosome 6). Hypermethylation occurred on STK19, a MHC class III gene for RNA surveillance [ , ]. Hypermethylation also occurred on a gene for preventing tumors (TNFB) [ ] and a gene for responding to antigen-antibody complexes (C2). Polymorphisms in HLA-DMB antigen and SAPCD1, another class III MHC gene [ ], at chromosome 6p21.3 had links to Kaposi sarcoma [ ]. Human herpesvirus 8 (Kaposi sarcoma virus) is a Kaposi sarcoma driver and is closely related to EBV.These results reveal that EBV has been attacking human DNA during evolution. There is a piRNA defense mechanism for human DNA near critical immune system genes, but both EBV-associated cancers and breast cancers inactivate some of the genes that guard piRNA defenses. The histocompatibility antigen gene region of chromosome 6 can be extensively fragmented in EBV-associated and breast cancers. MHC genes have the largest number of polymorphic forms in the human genome. This variation creates differences in viral susceptibility and inactivation. Even though most people are infected, not everyone will get an EBV-related disease or cancer.
Viral Sequences in Human Genomes as Hypermutation and Rearrangement Sites in Breast Cancers
The next question was whether EBV or other virus-like sequences in the human genome cause multiple rearrangements and clustered hypermutations (chromothripsis). As shown in
, many positions on chromosome 6 where chromothripsis occurs [ ] congregated around virus sequence start positions. A total of 1090 genome coordinates described chromothripsis fragments with copy number ≥3. These coordinates were unlikely to be random since they did not follow a normal distribution (P<.001). By simple linear regression analysis (R2=0.93), many viral sequence coordinates strongly correlated with chromothripsis positions. shows that as you move further away from a chromothripsis breakpoint, the frequency of breast cancer homology (score>500) to viruses decreases. This result implicates viral sequences as preferred sites where breast cancer chromosomes begin to fall apart. The equation shown mathematically describes the relationship between chromothripsis frequency and distance from viral sequences, and the constant in the equation suggests a baseline level of breakpoints.These results suggest that homologous virus sequences at multiple positions could confuse DNA repairs already compromised by EBV in breast cancer and contribute to chromothripsis and clustered rearrangements.
EBV and Metastasis
The last question was whether EBV contributes to breast cancer metastasis. According to Yates et al [
], relapsed and metastatic breast cancer tumors keep their tumor-driver gene mutations and continue acquiring new ones. Late mutations in JAK-STAT and SWI-SNF signaling pathways drive established breast cancers into metastasis.NPC often loses type-1 interferon genes (IFNA1, IFNA2, IFNA8, and IFNE) and nearby MTAP (32%-34% [
]) by homozygous deletions at chromosome 9p21.3. Interferons initiate canonical JAK-STAT signaling by binding to cell surface receptors that then activate internal Janus kinases (JAKs). The activated JAKs phosphorylate cytoplasmic STAT (signal transducer and activator of transcription) proteins, which travel to the cell nucleus to activate interferon-responsive genes. The percentages of breast cancers on the COSMIC website with mutations in a “JAK” or “STAT” isoform or transcript variant were calculated: 7.8% had a JAK mutation and 36.7% had a STAT mutation. Deletions of interferon genes in NPC also facilitate viral replication and block interferon from activating JAK-STAT signaling. Breast cancers ( ) have 65 breakpoints strictly within this interferon-MTAP region (21,579,478‐20,503,534 on chromosome 9), not counting longer fragments that include the interval. As shown in , breast cancer breakpoints align well with EBV-associated cancer breakpoints near the large cluster of interferon genes on chromosome 9.Mutations in EBV-associated cancers show that Yates metastasis driver gene damage accompanies EBV infection. SWI-SNF (switch/sucrose non-fermentable) is a complex that repositions nucleosomes and supports genome stability [
]. SWI-SNF addresses obstacles to replication sensed by the FA-BRCA pathway [ , ]. Referring back to , clustered breast cancer breakpoints on chromosome 17 around EBV breakpoints affect the SWI-SNF component SMARCE1. In addition, breast cancers can disable SWI-SNF by targeting ARID genes [ ]. ARID1A is a COSMIC top-20 most frequently mutated gene in breast cancer. Like breast cancer, NPC has multiple recurrent aberrations in ARID1A genes. As shown in , ARID1A lies near a hot spot where multiple breast cancer breakpoints approximately aligned with breakage points in EBV-associated cancers. The loss of ARID1A activates Annexin A1, which aligned closely with a region targeted by EBV-associated cancers on chromosome 9. A chromosome-1 site at about 150 million bp had a nearby histone-rich gene region. Histones are chromatin structures that SWI-SNF dynamically remodels to regulate access to genetic information. Histones can profoundly affect metastasis [ ]. also reveals many additional alignments between breakpoints in breast and EBV-associated cancers that were not investigated further.NPC often inactivates SWI-SNF components BAP1 and PBRM1 within a frequently damaged 3p21.3 gene cluster [
] at 52,400,000‐53,000,000 on chromosome 3. Analyses of breast cancers found 18 breakpoints within this short interval. DLBCL, another EBV-linked cancer, also had recurrent alterations in components of SWI-SNF complexes [ ].The Warburg effect (oxidative glycolysis) [
] favors metastasis. The Warburg effect occurs in NPC because pyruvate dehydrogenase (PDHB) genes on chromosome 3p are deleted or rearranged in almost all cases. Similar changes to chromosome 3p were found in breast cancers, which also undergo the Warburg effect [ ]. This Warburg metabolic switch favors metastasis because it mitigates oxidative stress on cancer cells. Large amounts of lactate accumulate in the absence of PDHB to acidify the tumor microenvironment and interfere with the destruction of metastatic cells [ ].This section’s results show that EBV may push breast cancer into metastasis by interfering with JAK-STAT and SWI-SNF signaling pathways to facilitate viral replication while making the microenvironment more favorable to tumor growth.
Alternative Explanations for Breast Cancer Breakpoints That Do Not Involve EBV Variants
Subgroups
To determine whether breakpoint similarities in viral and breast cancers depended on specific subgroups, relationships to NPC were compared in triple-negative and HER2-positive breast cancers (20 and 22 patients, respectively). Triple-negative breast cancers are likely to be BRCA1 mutation positive [
], while HER2 amplification is uncommon in BRCA1 and BRCA2 mutation carriers [ ]. Although subgroup differences are noticeable, results still show that both subgroups had breakpoints on all chromosomes related to NPC (Figure S1A in ).Tumor-Infiltrating Lymphocytes
Tumor-infiltrating lymphocytes (TILs) are biomarkers for predicting breast cancer prognosis [
, ]. To test whether TILs cause chromosome breaks, breakpoint numbers in 16 breast cancers with severe lymphocyte infiltration were compared to 17 breast cancers with nil lymphocyte infiltration. The 2-tailed Student t test could not reject the null hypothesis that the numbers of breakpoints were statistically identical (P=.70; Figure S1B in ). This result does not rule out differences in prognosis due to differences in lymphocyte infiltration.Retroviruses
Retrovirus contributions to structural variations were estimated using data from cancer in 38 different tissues [
]. Retrotransposons make relatively modest contributions to breast cancer compared to, say, esophageal or oral (gums) cancer ( ). EBV can transactivate endogenous retroviruses [ , , ]. DNA near some breast cancer breakpoints resembles porcine endogenous retrovirus, HERV, and HIV1 (eg, ). The human genome also contains DNA matching the retrovirus mouse mammary tumor virus [ , ] at 23 sites that give BLAST homology scores>200. HPV variants are DNA viruses that are also implicated in breast cancer. HPVs were not assessed further, but they occasionally matched DNA near breast cancer breakpoints.Common Fragile Sites
Common fragile sites are site-specific breaks seen on metaphase chromosomes after inhibiting DNA synthesis via DNA polymerase inhibitors. Some common fragile sites [
] aligned with breast cancer breaks on chromosome 1, but breakpoints on most other chromosomes were incompatible. Chromosomes 8, 9, 11‐15, 17-19, 21, and 22 do not have common fragile sites but still have many breast cancer breaks [ ]. However, the human genome has over 13 million palindromes that are ≤40 bp [ ]. The generation of rare fragile sites by palindromes or their attraction to EBV cannot be excluded.Imperfect Palindrome Repeats
An alternative explanation for EBV-related carcinogenesis involves the docking of EBNA1 virus-tethering protein at imperfect palindromes [
] tandemly repeated on chromosome 11. The docked EBNA1 binds EBV circular episomes, and chromosome 11 breaks initiate malignancy. To test this explanation, existing literature data were first compared to the specific human EBNA1-binding site [ , , ]. The results (Table S2 in ) are incompatible with a single host sequence binding EBNA1.BLAST analysis showed that matches to the imperfect palindrome were likely due to pure chance with E values between 16 and 964 for 4352 matches, from 12 to 18 bp. Chromosome 11 had only 197 of these 4352 matches, and none were near the palindromic region. The prototype DNA palindrome (Table S2 in
, line 2) produced 7074 matches with E values ranging from 0.25 to 964. Further BLAST analyses of the slightly different docking sequence in EBNA1-DNA crystals (Table S3 in , line 1) against other genome assemblies [ ] revealed matches on chromosomes 2, 19, 4, and 12. Various isolates of HIVs had 52 matching sequences.In 94 BL samples from patients who were EBV positive, breakpoints concentrated within chromosomes 2, 8, 13, 14, and 22 (Figure S1D in
). Chromosome 14 contained 610 breakpoints (IgVH regions), and chromosome 2 (IgVK regions) contained 522 breakpoints. EBV hijacks activation-induced cytidine deaminase, a mutagenic enzyme that generates antibody gene variants in response to myriad antigens. In the 94 EBV-positive BL cases, the palindromic locus was nearly 100 million bp away from the principal breakpoint coordinates (Figure S1E in ). Only 19 (20%) of the 94 patients who were EBV positive [ ] had breakpoints anywhere on chromosome 11. The palindromic locus was also not involved in diverse cancers from 8227 patients [ ] ( ).The results in this section show that alternative explanations that invoke subgroups, TILs, retroviruses, or a specific palindromic repeat locus are incompatible with the associations between EBV-associated and breast cancers .
Discussion
Principal Findings
This study finds that EBV contributes to breast cancer by disabling safeguards against tumors. Cancer then occurs because the safeguards remain disabled even if the virus is cleared. Multiple independent analyses identified residual genetic and epigenetic damage in cancer genomes and formed the basis of the model in
. Breakpoints in breast cancers in high-risk women, sporadic breast cancers, and even ovarian cancers cluster around breakpoints in known EBV-related cancers, including NPC, BL, DLBCL, and GC. Some genes clustered near breakpoints in these diverse EBV-associated cancers are critical to preventing breast cancers. Some breast cancer breakpoints are near genes at EBV-docking sites. Varying numbers of DNA breaks occur within the highly polymorphic forms of MHC region genes on chromosome 6. This damage adds to susceptible polymorphisms and immunodeficiencies to help explain why not everyone develops EBV-related cancers. Near the MHC region on chromosome 6, piRNA sequences are regularly interspaced between viral DNA sequences. The sandwiched arrangements are presumptive evidence of past infection and probably represent a DNA defense mechanism. These defenses fail when chromosome 6 breaks apart near start points of the large number of repetitive viral sequences in the human genome. The viral sequences confuse repairs already damaged by EBV, and bursts of mutation occur where scrambled fragments ligate. EBV disables the most reliable restoration of broken chromosomes back to their native forms, so repairs form structures with multiple centromeres. These structures undergo additional rounds of fragmentation during cell division. The process continually forms new cancer driver mutations and allows cancer to come back after successful therapy ( ). An EBV methylation signature on chromosome 6 was far more abundant in 1538 breast cancers than in normal controls. Finally, EBV facilitates its own replication by damaging JAK-STAT and SWI-SNF signaling pathways, which pushes breast cancer into metastasis, while virus-associated changes on chromosome 3p interfere with the destruction of metastatic cells. Models [ , ] of EBV-infected human mammary cell cultures transplanted into immunosuppressed mice and EBV loss from NPC cells are consistent with these results.The study herein has current and future clinical implications in addressing cancers and chronic diseases. An early childhood vaccine against EBV may reduce the incidence of breast cancer on a global scale. If this vaccine even approaches the effectiveness of the HPV vaccine for cervical cancer, then the reduction of breast cancer incidence would be substantial. In breast cancer cases where active infection can be demonstrated, immunotherapy or antivirals can be considered. The results also heighten concern about hidden dangers from viral infections. EBV infection leaves behind persistent genome abnormalities (“long EBV”) linked to breast cancer. Not everyone develops an EBV-related cancer even though almost everyone is infected, suggesting risk assessment should include MHC polymorphisms. MHC genes have abundant connections to both EBV infection [
] and breast cancer [ - ].The strategy of using bioinformatics to identify markers of “long EBV” may well work for other cancers, multiple sclerosis [
], and other chronic diseases that are currently unexplained. Testing for persistent viral damage in genomes from biopsies is a new method for screening for breast cancer risk. The results may inform further prevention and treatment decisions. Cancer drug therapy has focused on finding and destroying cancer-driver gene products. The drugs are initially effective, sometimes for long periods, but then stop working. The cycles represented in are an occult, underlying process that can now be evaluated. Cancer treatment generates new clones that do not exist in the original population [ ]. The underlying genome damage and EBV scars continually produce new cancer-driver mutations. Some antigens targeted by successful therapy for hematologic malignancies [ ], such as DLBCL, may also be effective for breast cancers. The idea that breast cancers and hematologic malignancies can have similar breakpoints and translocation fusions suggests that there may be many more susceptible targets and that there are options to overcome resistance or tolerance [ ]. The findings may further stimulate research into other EBV-associated diseases and cancers, leading to better and broader understanding.Estrogen has been thought to generate the initial chromosome breakpoints leading to translocations in human breast cancer. However, young boys with BL do not produce estrogen from ovaries, yet
shows that their malignant B-cells have many breakpoints [ , ] that approximately match breast cancer breakpoints. Normally, aromatase catalyzes the rate-limiting step in estrogen production [ ], and aromatase acting on androgens is the primary source of most estrogens in breast tissue [ ]. EBV-infected cells lose control of aromatase activity [ ]. An EBV-mediated increase in aromatase activity explains why locations of breakpoints ( ) are relatively independent of estrogen receptor status in breast cancer [ ] and resemble locations in lymphoid cells ( - and ). Transcription in response to artificially high estrogen levels created by EBV then induces topoisomerase-mediated DNA breaks. Double-strand break repair genes remove topoisomerase from these complexes, but damage to this process leaves pathological enzyme complexes still bound at a DNA breakpoint [ - ]. As shown in , topoisomerase itself may be damaged. In either case, EBV-mediated deregulation of estrogen production, topoisomerase activity, and deaminases then collaborate to cause chromosome breaks and drive breast cancer.Breast cancer chromosome breakpoints cluster around genes near EBV-binding sites (
), further suggesting that EBV participates in causing the breaks. The breaks lead to pathogenic chromosome rearrangements because EBV-induced damage forces restoration into error-prone methods by suppressing FA-BRCA pathway intermediates [ , ]. Repairs using the FA-BRCA pathway [ ] need chromatin access, which requires the SMC5/6 cohesin complex [ , ]. In one scenario shown in , SMC5/6 interacts with a crucial pathway intermediate, the FANCD2-FANCI heterodimer (“D2-I”) [ , ]. EBV variants deplete SMC5/6, preventing FA-BRCA–mediated DNA repairs and leading to chromosomes with too many centromeres. When mitosis pulls apart multicentromere chromosome structures, the forces shatter the chromosome and induce mutation storms [ ]. EBV thus threatens a sprawling, interconnected repair system, including the BRCA pathway, FA proteins, an SMC5/6 scaffold, JAK-STAT signaling, and the SWI-SNF chromatin remodeling complex ( ).Of course, other environmental, genetic, or lifestyle factors also participate in breast cancer development, but EBV infection exacerbates their effects. Genome deficits in EBV-associated cancers and breast cancers interfere with restoring chromosomes from damage due to natural processes and exogenous mutagens. Some of this damage requires repair pathways that are subject to EBV interference.
Evidence underlying the model in
has independent support from the literature. For example, viral load is a marker for the extent of cell-free DNA fragmentation [ ]. EBV-mediated transformation routinely generates abnormal karyotypes [ ]. The binding of EBNA1 sequence variants increases NPC risk and drives EBV lytic gene expression [ , ], which requires EBV-encoded nucleases [ - ]. Other herpesviruses related to EBV share the ability to fragment DNA and subvert DNA repair pathways [ - ]. EBV facilitates its own replication by interfering with signaling pathways that prevent metastasis [ , - ]. Independent literature supports EBV participation in metastasis and the results shown in . NPC has the highest metastatic rate among all head and neck cancers, and the levels of circulating EBV markers are highly predictive [ ]. Finding EBV in lymph nodes of patients with NPC or primary cancer at an unknown site helps detect metastasis [ ]. NPC patients with ≥500 copies of EBV per mL plasma had significantly higher rates of liver metastasis than patients with lower EBV levels [ ]. EBV-infected B-cells and breast cancer cells both have amplified centrosomes ( ), the mitosis-organizing centers that exert structural control over cell division. The EBV protein thymidine kinase takes up residence in the centrosome [ ], and another EBV protein, BNRF1, initiates centrosome amplification in infected B-cells [ ]. Overduplication of centrosomes confuses chromatid attachments to spindle fibers during mitosis. Chromosomes do not distribute properly into daughter cells, creating mistakes when the genome replicates [ , ]. Neither centrosome amplification nor chromosome fragmentation (chromothripsis) requires large numbers of viral particles or active infection.Further bioinformatic tests may still add significant additional information. EBV activation brings massive changes to host chromatin methylation and structure [
, , ]. Breast cancers have hundreds of these changes [ ]. Results here further implicate epigenetic effects, so EBV effects on breast cancer epigenetics should be explored in more detail. EBV is implicated in cancers in multiple additional organs, and the methods developed here may help clarify its potential contributions. Predictions based on virus-human interaction structural biology may also be helpful. The ultimate direct test will be whether childhood recipients of an anti-EBV vaccine have reduced breast cancer incidence. If it even approaches the reduction of cervical cancer achieved by the HPV vaccine (up to 94%), a childhood EBV vaccine could effectively prevent many cases of breast cancer.Limitations
EBV itself creates a limitation because the virus can disappear after causing pathogenic genome damage that allows breast cancer to develop. This transitory virus presence forces the use of bioinformatics to look for persistent genome damage EBV leaves behind. EBV disappearance questions whether a group of cancers with EBV connections also contains “sporadic” cancers typed as EBV negative. The EBV-negative forms may have merely lost the criteria used to identify EBV infection, but EBV-related genome damage may still remain. Another limitation is that compared to breast cancers, known EBV-linked cancers such as GC, BL, and NPC are less common, so genome sequence data are also less common.
Conclusions
In summary, early childhood immunizations against inactivated EBV or selected EBV gene products may significantly reduce the incidence of breast, ovarian, and other cancers, and potentially unexplained chronic diseases. EBV variants lead to DNA breaks, mitotic abnormalities, and the loss of safeguards that protect against breast cancer and its metastasis. Breast cancer breakpoints cluster around breakpoints in EBV cancers, disrupting genes essential to prevent viral infection and breast cancers. A CRISPR-like region on chromosome 6 sequesters some of the thousands of pieces of EBV sequences in the human genome. The same area of chromosome 6 undergoes variable damage in breast cancer, contributing to the reason not everyone with EBV infection develops cancer. In susceptible people, EBV infection leaves behind pathogenic cancer-associated genome abnormalities (“long EBV”). Clinical implications include improvements in evaluating the chances that cancer will return, increased use of immunotherapy for patients with breast cancer that have active infection, and greater urgency in developing an effective EBV vaccine,.
Data Availability
The primary dataset and calculations that were generated or analyzed during this study are included. Datasets not included are freely available from the original sources or the corresponding author on reasonable request.
Conflicts of Interest
None declared.
Glossary and abbreviations.
DOCX File, 20 KBMost of the calculations used in this work.
XLSX File, 7644 KBThe exact distances between nasopharyngeal cancer and breast cancer breakpoints on chromosome 1. These breaks gather around a few low valleys that periodically occur across the whole chromosome, but the data points are too numerous to display, making the results difficult to interpret.
PNG File, 213 KBGene functions at breast cancer breakpoints that clustered around breakpoints in EBV-associated cancers (GC, BL, and NPC), and EBNA1-binding sequences reported in the human genome. BL: Burkitt lymphoma; EBNA1: Epstein-Barr virus nuclear antigen 1; EBV: Epstein-Barr virus; NPC: nasopharyngeal cancer.
DOCX File, 31 KBAlternative explanations.
PNG File, 374 KBReferences
- DeSantis CE, Ma J, Gaudet MM, et al. Breast cancer statistics, 2019. CA Cancer J Clin. Nov 2019;69(6):438-451. [CrossRef] [Medline]
- QuickStats: age-adjusted death rates* for female breast cancer,† by state — National Vital Statistics System, United States, 2019§. MMWR Morb Mortal Wkly Rep. Oct 1, 2021;70(39):1391. [CrossRef] [Medline]
- Sarid R, Gao SJ. Viruses and human cancer: from detection to causality. Cancer Lett. Jun 28, 2011;305(2):218-227. [CrossRef] [Medline]
- zur Hausen H. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer. May 2002;2(5):342-350. [CrossRef] [Medline]
- Balfour HHJ, Sifakis F, Sliman JA, Knight JA, Schmeling DO, Thomas W. Age-specific prevalence of Epstein–Barr virus infection among individuals aged 6–19 years in the United States and factors affecting its acquisition. J Infect Dis. Oct 15, 2013;208(8):1286-1293. [CrossRef] [Medline]
- Huo Q, Zhang N, Yang Q. Epstein-Barr virus infection and sporadic breast cancer risk: a meta-analysis. PLoS One. 2012;7(2):e31656. [CrossRef] [Medline]
- Fina F, Romain S, Ouafik L, et al. Frequency and genome load of Epstein-Barr virus in 509 breast cancers from different geographical areas. Br J Cancer. Mar 23, 2001;84(6):783-790. [CrossRef] [Medline]
- Hu H, Luo ML, Desmedt C, et al. Epstein-Barr virus infection of mammary epithelial cells promotes malignant transformation. EBioMedicine. Jul 2016;9:148-160. [CrossRef] [Medline]
- God JM, Haque A. Burkitt lymphoma: pathogenesis and immune evasion. J Oncol. 2010;2010:516047. [CrossRef] [Medline]
- Xu M, Yao Y, Chen H, et al. Genome sequencing analysis identifies Epstein-Barr virus subtypes associated with high risk of nasopharyngeal carcinoma. Nat Genet. Jul 2019;51(7):1131-1136. [CrossRef] [Medline]
- Bruce JP, To KF, Lui VWY, et al. Whole-genome profiling of nasopharyngeal carcinoma reveals viral-host co-operation in inflammatory NF-κB activation and immune escape. Nat Commun. Jul 7, 2021;12(1):4193. [CrossRef] [Medline]
- Fountzilas G, Psyrri A, Giannoulatou E, et al. Prevalent somatic BRCA1 mutations shape clinically relevant genomic patterns of nasopharyngeal carcinoma in Southeast Europe. Int J Cancer. Jan 1, 2018;142(1):66-80. [CrossRef] [Medline]
- Devanaboyina M, Kaur J, Whiteley E, et al. NF-κB signaling in tumor pathways focusing on breast and ovarian cancer. Oncol Rev. Oct 3, 2022;16:10568. [CrossRef] [Medline]
- Lung RWM, Tong JHM, Ip LM, et al. EBV-encoded miRNAs can sensitize nasopharyngeal carcinoma to chemotherapeutic drugs by targeting BRCA1. J Cell Mol Med. Nov 2020;24(22):13523-13535. [CrossRef] [Medline]
- Hau PM, Tsao SW. Epstein-Barr virus hijacks DNA damage response transducers to orchestrate its life cycle. Viruses. Nov 16, 2017;9(11):341. [CrossRef] [Medline]
- Dheekollu J, Wiedmer A, Ayyanathan K, Deakyne JS, Messick TE, Lieberman PM. Cell-cycle-dependent EBNA1-DNA crosslinking promotes replication termination at oriP and viral episome maintenance. Cell. Feb 4, 2021;184(3):643-654.e13. [CrossRef] [Medline]
- Rossi F, Helbling-Leclerc A, Kawasumi R, et al. SMC5/6 acts jointly with Fanconi anemia factors to support DNA repair and genome stability. EMBO Rep. Feb 5, 2020;21(2):e48222. [CrossRef] [Medline]
- Yiu SPT, Guo R, Zerbe C, Weekes MP, Gewurz BE. Epstein-Barr virus BNRF1 destabilizes SMC5/6 cohesin complexes to evade its restriction of replication compartments. Cell Rep. Mar 8, 2022;38(10):110411. [CrossRef] [Medline]
- Fan Y, Ying H, Wu X, et al. The mutational pattern of homologous recombination (HR)-associated genes and its relevance to the immunotherapeutic response in gastric cancer. Cancer Biol Med. Nov 15, 2020;17(4):1002-1013. [CrossRef] [Medline]
- Parvin S, Labrada AR, Santiago GE, et al. Novel role of LMO2 in DNA repair control in diffuse large B cell lymphoma. Blood. Dec 2, 2016;128(22):776-776. [CrossRef]
- Busch K, Keller T, Fuchs U, et al. Identification of two distinct MYC breakpoint clusters and their association with various IGH breakpoint regions in the t(8;14) translocations in sporadic Burkitt-lymphoma. Leukemia. Aug 2007;21(8):1739-1751. [CrossRef] [Medline]
- Chong LC, Ben-Neriah S, Slack GW, et al. High-resolution architecture and partner genes of MYC rearrangements in lymphoma with DLBCL morphology. Blood Adv. Oct 23, 2018;2(20):2755-2765. [CrossRef] [Medline]
- Xu J, Chen Y, Olopade OI. MYC and breast cancer. Genes Cancer. Jun 2010;1(6):629-640. [CrossRef] [Medline]
- Umbreit NT, Zhang CZ, Lynch LD, et al. Mechanisms generating cancer genome complexity from a single cell division error. Science. Apr 17, 2020;368(6488):eaba0712. [CrossRef] [Medline]
- McClintock B. The stability of broken ends of chromosomes in Zea mays. Genetics. Mar 1941;26(2):234-282. [CrossRef] [Medline]
- Lee JJK, Jung YL, Cheong TC, et al. ERα-associated translocations underlie oncogene amplifications in breast cancer. Nature New Biol. Jun 2023;618(7967):1024-1032. [CrossRef] [Medline]
- McAulay KA, Higgins CD, Macsween KF, et al. HLA class I polymorphisms are associated with development of infectious mononucleosis upon primary EBV infection. J Clin Invest. Oct 2007;117(10):3042-3048. [CrossRef] [Medline]
- Aboulaghras S, Khalid A, Makeen HA, et al. Polymorphism of HLA and susceptibility of breast cancer. Front Biosci (Landmark Ed). Feb 5, 2024;29(2):55. [CrossRef] [Medline]
- Yates LR, Knappskog S, Wedge D, et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell. Aug 14, 2017;32(2):169-184.e7. [CrossRef] [Medline]
- Friedenson B. Dewey defeats Truman and cancer statistics. J Natl Cancer Inst. Aug 19, 2009;101(16):1157. [CrossRef] [Medline]
- Nik-Zainal S, Davies H, Staaf J, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature New Biol. Jun 2, 2016;534(7605):47-54. [CrossRef] [Medline]
- Staaf J, Glodzik D, Bosch A, et al. Whole-genome sequencing of triple-negative breast cancers in a population-based clinical study. Nat Med. Oct 2019;25(10):1526-1533. [CrossRef] [Medline]
- Nones K, Johnson J, Newell F, et al. Whole-genome sequencing reveals clinically relevant insights into the aetiology of familial breast cancers. Ann Oncol. Jul 1, 2019;30(7):1071-1079. [CrossRef] [Medline]
- Batra RN, Lifshitz A, Vidakovic AT, et al. DNA methylation landscapes of 1538 breast cancers reveal a replication-linked clock, epigenomic instability and cis-regulation. Nat Commun. Sep 13, 2021;12(1):5406. [CrossRef] [Medline]
- Cortés-Ciriano I, Lee JJK, Xi R, et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat Genet. Mar 2020;52(3):331-341. [CrossRef] [Medline]
- Petrucelli N, Daly MB, Pal T. BRCA1- and BRCA2-associated hereditary breast and ovarian cancer. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, editors. GeneReviews. University of Washington, Seattle; 1993. URL: https://www.ncbi.nlm.nih.gov/books/NBK1247/ [Accessed 2025-01-09]
- Lalloo F, Varley J, Moran A, et al. BRCA1, BRCA2 and TP53 mutations in very early-onset breast cancer with associated risks to relatives. Eur J Cancer. May 2006;42(8):1143-1150. [CrossRef] [Medline]
- Joos S, Falk MH, Lichter P, et al. Variable breakpoints in Burkitt lymphoma cells with chromosomal t(8;14) translocation separate c-myc and the IgH locus up to several hundred kb. Hum Mol Genet. Nov 1992;1(8):625-632. [CrossRef] [Medline]
- Joos S, Haluska FG, Falk MH, et al. Mapping chromosomal breakpoints of Burkitt’s t(8;14) translocations far upstream of c-myc. Cancer Res. Dec 1, 1992;52(23):6547-6552. [Medline]
- López C, Kleinheinz K, Aukema SM, et al. Genomic and transcriptomic changes complement each other in the pathogenesis of sporadic Burkitt lymphoma. Nat Commun. Mar 29, 2019;10(1):1459. [CrossRef] [Medline]
- Xing R, Zhou Y, Yu J, et al. Whole-genome sequencing reveals novel tandem-duplication hotspots and a prognostic mutational signature in gastric cancer. Nat Commun. May 2, 2019;10(1):2037. [CrossRef] [Medline]
- Nangalia J, Campbell PJ. Genome sequencing during a patient’s journey through cancer. N Engl J Med. Nov 28, 2019;381(22):2145-2156. [CrossRef] [Medline]
- Mount DW. Using the Basic Local Alignment Search Tool (BLAST). CSH Protoc. Jul 1, 2007;2007(7):pdb.top17. [CrossRef] [Medline]
- Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol. Oct 5, 1990;215(3):403-410. [CrossRef] [Medline]
- Zhang Z, Schwartz S, Wagner L, Miller W. A greedy algorithm for aligning DNA sequences. J Comput Biol. 2000;7(1-2):203-214. [CrossRef] [Medline]
- Rangwala SH, Kuznetsov A, Ananiev V, et al. Accessing NCBI data using the NCBI Sequence Viewer and Genome Data Viewer (GDV). Genome Res. Jan 2021;31(1):159-169. [CrossRef] [Medline]
- Kim KD, Tanizawa H, de Leo A, et al. Epigenetic specifications of host chromosome docking sites for latent Epstein-Barr virus. Nat Commun. Feb 13, 2020;11(1):877. [CrossRef] [Medline]
- Lu F, Wikramasinghe P, Norseen J, et al. Genome-wide analysis of host-chromosome binding sites for Epstein-Barr virus nuclear antigen 1 (EBNA1). Virol J. Oct 7, 2010;7:262. [CrossRef] [Medline]
- Sai Lakshmi S, Agrawal S. piRNABank: a web resource on classified and clustered Piwi-interacting RNAs. Nucleic Acids Res. Jan 2008;36(Database issue):D173-D177. [CrossRef] [Medline]
- Wang J, Zhang P, Lu Y, et al. piRBase: a comprehensive database of piRNA sequences. Nucleic Acids Res. Jan 8, 2019;47(D1):D175-D180. [CrossRef] [Medline]
- Tang MH, Varadan V, Kamalakaran S, Zhang MQ, Dimitrova N, Hicks J. Major chromosomal breakpoint intervals in breast cancer co-localize with differentially methylated regions. Front Oncol. Dec 27, 2012;2:197. [CrossRef] [Medline]
- Mann HB, Whitney DR. On a test of whether one of two random variables is stochastically larger than the other. Ann Math Statist. Mar 1947;18(1):50-60. [CrossRef]
- Shapiro SS, Wilk MB. An analysis of variance test for normality (complete samples). Biometrika. Dec 1965;52(3/4):591-611. [CrossRef]
- Kumar R, Nagpal G, Kumar V, Usmani SS, Agrawal P, Raghava GPS. HumCFS: a database of fragile sites in human chromosomes. BMC Genomics. Apr 18, 2019;19(Suppl 9):985. [CrossRef] [Medline]
- Maccaroni K, Balzano E, Mirimao F, Giunta S, Pelliccia F. Impaired replication timing promotes tissue-specific expression of common fragile sites. Genes (Basel). Mar 19, 2020;11(3):326. [CrossRef] [Medline]
- European Commission: Directorate-General for Research and Innovation. Improving access to and reuse of research results, publications and data for scientific purposes – study to evaluate the effects of the EU copyright framework on research and the effects of potential interventions and to identify and present relevant provisions for research in EU data and digital legislation, with a focus on rights and obligations. Publications Office of the European Union; 2024. URL: https://data.europa.eu/doi/10.2777/633395 [Accessed 2025-01-09]
- Zheng B, Liu XL, Fan R, et al. The landscape of cell-free HBV integrations and mutations in cirrhosis and hepatocellular carcinoma patients. Clin Cancer Res. Jul 1, 2021;27(13):3772-3783. [CrossRef] [Medline]
- Foster KA, Harrington P, Kerr J, et al. Somatic and germline mutations of the BRCA2 gene in sporadic ovarian cancer. Cancer Res. Aug 15, 1996;56(16):3622-3625. [Medline]
- Friedenson B. The BRCA1/2 pathway prevents hematologic cancers in addition to breast and ovarian cancers. BMC Cancer. Aug 6, 2007;7:152. [CrossRef] [Medline]
- Friedenson B. Comment on 'The incidence of leukaemia in women with BRCA1 and BRCA2 mutations: an International Prospective Cohort Study'. Br J Cancer. Aug 23, 2016;115(5):e2. [CrossRef] [Medline]
- Parvin S, Ramirez-Labrada A, Aumann S, et al. LMO2 confers synthetic lethality to PARP inhibition in DLBCL. Cancer Cell. Sep 16, 2019;36(3):237-249.e6. [CrossRef] [Medline]
- Compagno M, Lim WK, Grunn A, et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. Jun 4, 2009;459(7247):717-721. [CrossRef] [Medline]
- Ceribelli M, Kelly PN, Shaffer AL, et al. Blockade of oncogenic IκB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors. Proc Natl Acad Sci U S A. Aug 5, 2014;111(31):11365-11370. [CrossRef] [Medline]
- Montes-Moreno S, Odqvist L, Diaz-Perez JA, et al. EBV-positive diffuse large B-cell lymphoma of the elderly is an aggressive post-germinal center B-cell neoplasm characterized by prominent nuclear factor-kB activation. Mod Pathol. Jul 2012;25(7):968-982. [CrossRef] [Medline]
- Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. Mar 15, 2018;555(7696):321-327. [CrossRef] [Medline]
- Li JSZ, Abbasi A, Kim DH, Lippman SM, Alexandrov LB, Cleveland DW. Chromosomal fragile site breakage by EBV-encoded EBNA1 at clustered repeats. Nature. Apr 2023;616(7957):504-509. [CrossRef] [Medline]
- Hui KF, Chan TF, Yang W, et al. High risk Epstein‐Barr virus variants characterized by distinct polymorphisms in the EBER locus are strongly associated with nasopharyngeal carcinoma. Int J Cancer. Jun 15, 2019;144(12):3031-3042. [CrossRef] [Medline]
- Friedenson B. Evidence of lesions from Epstein-Barr virus infection in human breast cancer genomes. medRxiv. Preprint posted online on Jun 26, 2024. [CrossRef]
- Nurk S, Koren S, Rhie A, et al. The complete sequence of a human genome. Science. Apr 2022;376(6588):44-53. [CrossRef] [Medline]
- Luo WJ, Takakuwa T, Ham MF, et al. Epstein-Barr virus is integrated between REL and BCL-11A in American Burkitt lymphoma cell line (NAB-2). Lab Invest. Sep 2004;84(9):1193-1199. [CrossRef] [Medline]
- Tsao SW, Tsang CM, Lo KW. Epstein-Barr virus infection and nasopharyngeal carcinoma. Philos Trans R Soc Lond B Biol Sci. Oct 19, 2017;372(1732):20160270. [CrossRef] [Medline]
- Scott RS. Epstein-Barr virus: a master epigenetic manipulator. Curr Opin Virol. Oct 2017;26:74-80. [CrossRef] [Medline]
- Schott G, Garcia-Blanco MA. MHC class III RNA binding proteins and immunity. RNA Biol. May 2021;18(5):640-646. [CrossRef] [Medline]
- Zhou D, Lai M, Luo A, Yu CY. An RNA metabolism and surveillance quartet in the major histocompatibility complex. Cells. Aug 30, 2019;8(9):1008. [CrossRef] [Medline]
- Fernandes MT, Dejardin E, dos Santos NR. Context-dependent roles for lymphotoxin-β receptor signaling in cancer development. Biochim Biophys Acta. Apr 2016;1865(2):204-219. [CrossRef] [Medline]
- Shiina T, Blancher A, Inoko H, Kulski JK. Comparative genomics of the human, macaque and mouse major histocompatibility complex. Immunology. Feb 2017;150(2):127-138. [CrossRef] [Medline]
- Aissani B, Boehme AK, Wiener HW, Shrestha S, Jacobson LP, Kaslow RA. SNP screening of central MHC-identified HLA-DMB as a candidate susceptibility gene for HIV-related Kaposi’s sarcoma. Genes Immun. Sep 2014;15(6):424-429. [CrossRef] [Medline]
- Zhuang J, Huo Q, Yang F, Xie N. Perspectives on the role of histone modification in breast cancer progression and the advanced technological tools to study epigenetic determinants of metastasis. Front Genet. Oct 29, 2020;11:603552. [CrossRef] [Medline]
- Bayona-Feliu A, Barroso S, Muñoz S, Aguilera A. The SWI/SNF chromatin remodeling complex helps resolve R-loop-mediated transcription–replication conflicts. Nat Genet. Jul 2021;53(7):1050-1063. [CrossRef] [Medline]
- Harrod A, Lane KA, Downs JA. The role of the SWI/SNF chromatin remodelling complex in the response to DNA double strand breaks. DNA Repair (Amst). Sep 2020;93:102919. [CrossRef] [Medline]
- Andrades A, Peinado P, Alvarez-Perez JC, et al. SWI/SNF complexes in hematological malignancies: biological implications and therapeutic opportunities. Mol Cancer. Feb 21, 2023;22(1):39. [CrossRef] [Medline]
- Lu J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. Jun 2019;38(1-2):157-164. [CrossRef] [Medline]
- Chen H, Wu J, Zhang Z, et al. Association between BRCA status and triple-negative breast cancer: a meta-analysis. Front Pharmacol. Aug 21, 2018;9:909. [CrossRef] [Medline]
- Evans DG, Lalloo F, Howell S, Verhoef S, Woodward ER, Howell A. Low prevalence of HER2 positivity amongst BRCA1 and BRCA2 mutation carriers and in primary BRCA screens. Breast Cancer Res Treat. Feb 2016;155(3):597-601. [CrossRef] [Medline]
- Locy H, Verhulst S, Cools W, et al. Assessing tumor-infiltrating lymphocytes in breast cancer: a proposal for combining immunohistochemistry and gene expression analysis to refine scoring. Front Immunol. Feb 11, 2022;13:794175. [CrossRef] [Medline]
- Takada K, Kashiwagi S, Asano Y, et al. Prediction of distant metastatic recurrence by tumor-infiltrating lymphocytes in hormone receptor-positive breast cancer. BMC Womens Health. May 29, 2021;21(1):225. [CrossRef] [Medline]
- Meier UC, Cipian RC, Karimi A, Ramasamy R, Middeldorp JM. Cumulative roles for Epstein-Barr virus, human endogenous retroviruses, and human herpes virus-6 in driving an inflammatory cascade underlying MS pathogenesis. Front Immunol. Nov 1, 2021;12:757302. [CrossRef] [Medline]
- Mameli G, Poddighe L, Mei A, et al. Expression and activation by Epstein Barr virus of human endogenous retroviruses-W in blood cells and astrocytes: inference for multiple sclerosis. PLoS One. 2012;7(9):e44991. [CrossRef] [Medline]
- Lawson JS, Glenn WK. Evidence for a causal role by mouse mammary tumour-like virus in human breast cancer. NPJ Breast Cancer. Nov 7, 2019;5:40. [CrossRef] [Medline]
- Dudley JP, Golovkina TV, Ross SR. Lessons learned from mouse mammary tumor virus in animal models. ILAR J. 2016;57(1):12-23. [CrossRef] [Medline]
- Li Y, Roberts ND, Wala JA, et al. Patterns of somatic structural variation in human cancer genomes. Nature. Feb 2020;578(7793):112-121. [CrossRef] [Medline]
- Ganapathiraju MK, Subramanian S, Chaparala S, Karunakaran KB. A reference catalog of DNA palindromes in the human genome and their variations in 1000 Genomes. Hum Genome Var. Nov 20, 2020;7(1):40. [CrossRef] [Medline]
- Rawlins DR, Milman G, Hayward SD, Hayward GS. Sequence-specific DNA binding of the Epstein-Barr virus nuclear antigen (EBNA-1) to clustered sites in the plasmid maintenance region. Cell. Oct 1985;42(3):859-868. [CrossRef] [Medline]
- Bochkarev A, Barwell JA, Pfuetzner RA, Bochkareva E, Frappier L, Edwards AM. Crystal structure of the DNA-binding domain of the Epstein-Barr virus origin-binding protein, EBNA1, bound to DNA. Cell. Mar 8, 1996;84(5):791-800. [CrossRef] [Medline]
- Altemose N, Logsdon GA, Bzikadze AV, et al. Complete genomic and epigenetic maps of human centromeres. Science. Apr 2022;376(6588):eabl4178. [CrossRef] [Medline]
- Grande BM, Gerhard DS, Jiang A, et al. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma. Blood. Mar 21, 2019;133(12):1313-1324. [CrossRef] [Medline]
- Kim TM, Xi R, Luquette LJ, Park RW, Johnson MD, Park PJ. Functional genomic analysis of chromosomal aberrations in a compendium of 8000 cancer genomes. Genome Res. Feb 2013;23(2):217-227. [CrossRef] [Medline]
- Dittmer DP, Hilscher CJ, Gulley ML, Yang EV, Chen M, Glaser R. Multiple pathways for Epstein-Barr virus episome loss from nasopharyngeal carcinoma. Int J Cancer. Nov 1, 2008;123(9):2105-2112. [CrossRef] [Medline]
- Li Q, Cohen JI. Epstein-Barr virus and the human leukocyte antigen complex. Curr Clin Microbiol Rep. Sep 2019;6(3):175-181. [CrossRef] [Medline]
- Lei PJ, Pereira ER, Andersson P, et al. Cancer cell plasticity and MHC-II–mediated immune tolerance promote breast cancer metastasis to lymph nodes. J Exp Med. Sep 4, 2023;220(9):e20221847. [CrossRef] [Medline]
- Park IA, Hwang SH, Song IH, et al. Expression of the MHC class II in triple-negative breast cancer is associated with tumor-infiltrating lymphocytes and interferon signaling. PLoS One. Aug 17, 2017;12(8):e0182786. [CrossRef] [Medline]
- Song IH, Kim YA, Heo SH, et al. The association of estrogen receptor activity, interferon signaling, and MHC class I expression in breast cancer. Cancer Res Treat. Oct 2022;54(4):1111-1120. [CrossRef] [Medline]
- Bjornevik K, Cortese M, Healy BC, et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science. Jan 21, 2022;375(6578):296-301. [CrossRef] [Medline]
- Aissa AF, Islam ABMMK, Ariss MM, et al. Single-cell transcriptional changes associated with drug tolerance and response to combination therapies in cancer. Nat Commun. Mar 12, 2021;12(1):1628. [CrossRef] [Medline]
- Cao Y, Efetov SK, He M, et al. Updated clinical perspectives and challenges of chimeric antigen receptor-T cell therapy in colorectal cancer and invasive breast cancer. Arch Immunol Ther Exp (Warsz). Aug 11, 2023;71(1):19. [CrossRef] [Medline]
- Upton R, Banuelos A, Feng D, et al. Combining CD47 blockade with trastuzumab eliminates HER2-positive breast cancer cells and overcomes trastuzumab tolerance. Proc Natl Acad Sci U S A. Jul 20, 2021;118(29):e2026849118. [CrossRef] [Medline]
- Burkhardt B, Michgehl U, Rohde J, et al. Clinical relevance of molecular characteristics in Burkitt lymphoma differs according to age. Nat Commun. Jul 6, 2022;13(1):3881. [CrossRef] [Medline]
- Dochi H, Kondo S, Murata T, et al. Estrogen induces the expression of EBV lytic protein ZEBRA, a marker of poor prognosis in nasopharyngeal carcinoma. Cancer Sci. Aug 2022;113(8):2862-2877. [CrossRef] [Medline]
- Zhu BT, Conney AH. Functional role of estrogen metabolism in target cells: review and perspectives. Carcinogenesis. Jan 1, 1998;19(1):1-27. [CrossRef] [Medline]
- Pommier Y, Nussenzweig A, Takeda S, Austin C. Human topoisomerases and their roles in genome stability and organization. Nat Rev Mol Cell Biol. Jun 2022;23(6):407-427. [CrossRef] [Medline]
- Sasanuma H, Tsuda M, Morimoto S, et al. BRCA1 ensures genome integrity by eliminating estrogen-induced pathological topoisomerase II-DNA complexes. Proc Natl Acad Sci U S A. Nov 6, 2018;115(45):E10642-E10651. [CrossRef] [Medline]
- Manguso N, Kim M, Joshi N, et al. TDP2 is a regulator of estrogen-responsive oncogene expression. NAR Cancer. Apr 8, 2024;6(2):zcae016. [CrossRef] [Medline]
- de Piccoli G, Cortes-Ledesma F, Ira G, et al. Smc5-Smc6 mediate DNA double-strand-break repair by promoting sister-chromatid recombination. Nat Cell Biol. Sep 2006;8(9):1032-1034. [CrossRef] [Medline]
- Tapia-Alveal C, Lin SJ, O’Connell MJ. Functional interplay between cohesin and Smc5/6 complexes. Chromosoma. Oct 2014;123(5):437-445. [CrossRef] [Medline]
- Irwan ID, Cullen BR. The SMC5/6 complex: an emerging antiviral restriction factor that can silence episomal DNA. PLoS Pathog. Mar 2, 2023;19(3):e1011180. [CrossRef] [Medline]
- Agashe S, Joseph CR, Reyes TAC, et al. Smc5/6 functions with Sgs1-Top3-Rmi1 to complete chromosome replication at natural pause sites. Nat Commun. Apr 8, 2021;12(1):2111. [CrossRef] [Medline]
- Truszewska A, Wirkowska A, Gala K, et al. EBV load is associated with cfDNA fragmentation and renal damage in SLE patients. Lupus. Jul 2021;30(8):1214-1225. [CrossRef] [Medline]
- Volleth M, Zenker M, Joksic I, Liehr T. Long-term culture of EBV-induced human lymphoblastoid cell lines reveals chromosomal instability. J Histochem Cytochem. Apr 2020;68(4):239-251. [CrossRef] [Medline]
- Dheekollu J, Malecka K, Wiedmer A, et al. Carcinoma-risk variant of EBNA1 deregulates Epstein-Barr virus episomal latency. Oncotarget. Jan 31, 2017;8(5):7248-7264. [CrossRef] [Medline]
- Buisson M, Géoui T, Flot D, et al. A bridge crosses the active-site canyon of the Epstein-Barr virus nuclease with DNase and RNase activities. J Mol Biol. Aug 28, 2009;391(4):717-728. [CrossRef] [Medline]
- Wu CC, Liu MT, Chang YT, et al. Epstein-Barr virus DNase (BGLF5) induces genomic instability in human epithelial cells. Nucleic Acids Res. Apr 2010;38(6):1932-1949. [CrossRef] [Medline]
- Chiu SH, Wu MC, Wu CC, et al. Epstein-Barr virus BALF3 has nuclease activity and mediates mature virion production during the lytic cycle. J Virol. May 2014;88(9):4962-4975. [CrossRef] [Medline]
- Chiu SH, Wu CC, Fang CY, et al. Epstein-Barr virus BALF3 mediates genomic instability and progressive malignancy in nasopharyngeal carcinoma. Oncotarget. Sep 30, 2014;5(18):8583-8601. [CrossRef] [Medline]
- O’Neill FJ, Miles CP. Chromosome changes in human cells induced by herpes simplex, types 1 and 2. Nature. Aug 23, 1969;223(5208):851-852. [CrossRef] [Medline]
- Mertens ME, Knipe DM. Herpes simplex virus 1 manipulates host cell antiviral and proviral DNA damage responses. mBio. Feb 9, 2021;12(1):e03552-20. [CrossRef] [Medline]
- Schumacher AJ, Mohni KN, Kan Y, Hendrickson EA, Stark JM, Weller SK. The HSV-1 exonuclease, UL12, stimulates recombination by a single strand annealing mechanism. PLoS Pathog. 2012;8(8):e1002862. [CrossRef] [Medline]
- Ezeonwumelu IJ, Garcia-Vidal E, Ballana E. JAK-STAT pathway: a novel target to tackle viral infections. Viruses. Nov 27, 2021;13(12):2379. [CrossRef] [Medline]
- Jangra S, Bharti A, Lui WY, et al. Suppression of JAK-STAT signaling by Epstein-Barr virus tegument protein BGLF2 through recruitment of SHP1 phosphatase and promotion of STAT2 degradation. J Virol. Sep 27, 2021;95(20):e0102721. [CrossRef] [Medline]
- Wu DY, Krumm A, Schubach WH. Promoter-specific targeting of human SWI-SNF complex by Epstein-Barr virus nuclear protein 2. J Virol. Oct 2000;74(19):8893-8903. [CrossRef] [Medline]
- Su MT, Wang YT, Chen YJ, Lin SF, Tsai CH, Chen MR. The SWI/SNF chromatin regulator BRG1 modulates the transcriptional regulatory activity of the Epstein-Barr virus DNA polymerase processivity factor BMRF1. J Virol. Apr 13, 2017;91(9):e02114-16. [CrossRef] [Medline]
- Nakao K, Yuge T, Mochiki M, Nibu KI, Sugasawa M. Detection of Epstein-Barr virus in metastatic lymph nodes of patients with nasopharyngeal carcinoma and a primary unknown carcinoma. Arch Otolaryngol Head Neck Surg. Mar 2003;129(3):338-340. [CrossRef] [Medline]
- Tao D, Zhang N, Huang Q, et al. Association of Epstein-Barr virus infection with peripheral immune parameters and clinical outcome in advanced nasopharyngeal carcinoma. Sci Rep. Dec 15, 2020;10(1):21976. [CrossRef] [Medline]
- Gill MB, Kutok JL, Fingeroth JD. Epstein-Barr virus thymidine kinase is a centrosomal resident precisely localized to the periphery of centrioles. J Virol. Jun 15, 2007;81(12):6523-6535. [CrossRef] [Medline]
- Shumilov A, Tsai MH, Schlosser YT, et al. Epstein-Barr virus particles induce centrosome amplification and chromosomal instability. Nat Commun. Feb 10, 2017;8:14257. [CrossRef] [Medline]
- Lingle WL, Lutz WH, Ingle JN, Maihle NJ, Salisbury JL. Centrosome hypertrophy in human breast tumors: implications for genomic stability and cell polarity. Proc Natl Acad Sci U S A. Mar 17, 1998;95(6):2950-2955. [CrossRef] [Medline]
- Buschle A, Mrozek-Gorska P, Cernilogar FM, et al. Epstein-Barr virus inactivates the transcriptome and disrupts the chromatin architecture of its host cell in the first phase of lytic reactivation. Nucleic Acids Res. Apr 6, 2021;49(6):3217-3241. [CrossRef] [Medline]
Abbreviations
BL: Burkitt lymphoma |
BLAST: Basic Local Alignment Search Tool |
bp: base pairs |
COSMIC: Catalog of Somatic Mutations in Cancer |
CRISPR: clustered regularly interspaced short palindromic repeats |
DLBCL: diffuse large B-cell lymphoma |
EBNA1: Epstein-Barr virus nuclear antigen 1 |
EBV: Epstein-Barr virus |
FA: Fanconi anemia |
GC: gastric cancer |
HER2: human epidermal growth factor receptor 2 |
HERV: human endogenous retrovirus |
HIV1: human immunodeficiency virus type 1 |
HLA: human leukocyte antigen |
HPV: human papillomavirus |
JAK: Janus kinase |
MHC: major histocompatibility complex |
NF-κB: nuclear factor–κB |
NPC: nasopharyngeal cancer |
piRNA: Piwi-interacting RNA |
SWI-SNF : switch/sucrose non-fermentable |
TIL: tumor-infiltrating lymphocyte |
Edited by Amy Schwartz; submitted 10.07.23; peer-reviewed by Anonymous, Anonymous; final revised version received 19.11.24; accepted 20.11.24; published 29.01.25.
Copyright© Bernard Friedenson. Originally published in JMIRx Med (https://med.jmirx.org), 29.1.2025.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in JMIRx Med, is properly cited. The complete bibliographic information, a link to the original publication on https://med.jmirx.org/, as well as this copyright and license information must be included.