Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Cancer ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801336

ABSTRACT

Cervical cancer screening programs, including triage tests, need redesigning as human papillomavirus (HPV)-vaccinated women are entering the programs. Methylation markers offer a potential solution to reduce false-positive rates by identifying clinically relevant cervical lesions with progressive potential. In a nested case-control study, 9242 women who received the three-dose HPV16/18-vaccine at ages 12-15 or 18 in a community-randomized trial were included. Subsequently, they were re-randomized for either frequent or infrequent cervical cancer screening trials. Over a 15-year post-vaccination follow-up until 2022, 17 high-grade squamous intraepithelial lesion (HSIL) and 15 low-grade (LSIL) cases were identified at the 25-year screening round, alongside 371 age and community-matched HPV16/18-vaccinated controls. Methylation analyses were performed on cervical samples collected at age 25, preceding histologically confirmed LSIL or HSIL diagnoses. DNA methylation of viral (HPV16/18/31/33) and host-cell genes (EPB41L3, FAM19A4, and miR124-2) was measured, along with HPV-genotyping. No HPV16/18 HSIL cases were observed. The predominant HPV-genotypes were HPV52 (29.4%), HPV59/HPV51/HPV58 (each 23.5%), and HPV33 (17.7%). Methylation levels were generally low, with no significant differences in mean methylation levels of viral or host-cell genes between the LSIL/HSIL and controls. However, a significant difference in methylation levels was found between HSIL cases and controls when considering a combination of viral genes and EPB41L3 (p value = .0001). HPV-vaccinated women with HSIL had HPV infections with uncommon HPV types that very rarely cause cancer and displayed low methylation levels. Further investigation is warranted to understand the likely regressive nature of HSIL among HPV-vaccinated women and its implications for management.

2.
PLoS Med ; 20(10): e1004304, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37889928

ABSTRACT

BACKGROUND: Cervical screening programs use testing for human papillomavirus (HPV) genotypes. Different HPV types differ greatly in prevalence and oncogenicity. We estimated the impact of cervical screening and follow-up for each HPV type. METHODS AND FINDINGS: For each type of HPV, we calculated the number of women needed to screen (NNS) and number of women needing follow-up (NNF) to detect or prevent one cervical cancer case, using the following individual level input data (i) screening and cancer data for all women aged 25 to 80 years, resident in Sweden during 2004 to 2011 (N = 3,568,938); (ii) HPV type-specific prevalences and screening histories among women with cervical cancer in Sweden in 2002 to 2011(N = 4,254); (iii) HPV 16/18/other HPV prevalences in the population-based HPV screening program (N = 656,607); and (iv) exact HPV genotyping in a population-based cohort (n = 12,527). Historical screening attendance was associated with a 72% reduction of cervical cancer incidence caused by HPV16 (71.6%, 95% confidence interval (CI) [69.1%, 73.9%]) and a 54% reduction of cancer caused by HPV18 (53.8%, 95% CI [40.6%, 63.1%]). One case of HPV16-caused cervical cancer could be prevented for every 5,527 women attending screening (number needed to screen, NNS). Prevention of one case of HPV16-caused cervical cancer required follow-up of 147 HPV16-positive women (number needed to follow-up, NNF). The NNS and NNF were up to 40 to 500 times higher for HPV types commonly screened for with lower oncogenic potential (HPV35,39,51,56,59,66,68). For women below 30 years of age, NNS and NNF for HPV16 were 4,747 and 289, respectively, but >220,000 and >16,000 for HPV35,39,51,56,59,66,68. All estimates were either age-standarized or age-stratified. The primary limitation of our study is that NNS is dependent on the HPV prevalence that can differ between populations and over time. However, it can readily be recalculated in other settings and monitored when HPV type-specific prevalence changes. Other limitations include that in some age groups, there was little data and extrapolations had to be made. Finally, there were very few cervical cancer cases associated with certain HPV types in young age group. CONCLUSIONS: In this study, we observed that the impact of cervical cancer screening varies depending on the HPV type screened for. Estimating and monitoring the impact of screening by HPV type can facilitate the design of effective and efficient HPV-based cervical screening programs. TRIAL REGISTRATION: ClinicalTrials.gov with numbers NCT00479375, NCT01511328.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/prevention & control , Early Detection of Cancer/methods , Human Papillomavirus Viruses , Human papillomavirus 16 , Papillomavirus Infections/diagnosis , Papillomavirus Infections/epidemiology , Papillomavirus Infections/complications , Human papillomavirus 18 , Papillomaviridae/genetics , Genotype
3.
J Med Virol ; 95(12): e29332, 2023 12.
Article in English | MEDLINE | ID: mdl-38115556

ABSTRACT

The International Human Papillomavirus (HPV) Reference Center (IHRC) confirms and assigns type numbers to novel HPV types, maintains a reference clone repository, and issues international proficiency panels for HPV screening and genotyping. Furthermore, the Center coordinates the Global HPV Reference Laboratory Network that promotes collaboration and international exchange of experiences among national HPV reference laboratories, to further international standardization and quality assurance in the HPV field. The established HPV types (n = 225) belong to 5 different genera: alpha (n = 65), beta (n = 54), gamma (n = 102), mu (n = 3) and nu (n = 1). Since the last published IHRC overview in 2018, 6 novel types have been established, with 5/6 belonging to the gamma genus and 1/6 to beta genus. Also, 474 reference clones have been provided to 55 different research laboratories and the global proficiency program for HPV genotyping has seen an increasing proficiency (despite a decrease seen in 2019), from 68% proficiency in 2017 to 77.3% in 2022. The first proficiency study for HPV screening found an international proficiency of up to 77%. In summary, increasing complexity of the HPVs and demands on quality assurance in the era of cervical cancer elimination requires international efforts to support proficiency and recognized quality and order among HPV types.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Human Papillomavirus Viruses , Papillomavirus Infections/diagnosis , Papillomaviridae/genetics , Reference Standards , Mass Screening , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/prevention & control
4.
J Infect Dis ; 226(7): 1195-1199, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35535025

ABSTRACT

For head-to-head comparison of human papillomavirus (HPV) antibody levels induced by different vaccines, 25-year-old vaccine-naive women were given either the bivalent (n = 188) or the nonavalent HPV vaccine (n = 184). Six months after vaccination antibodies against pseudovirions from 17 different HPV types (HPV6/11/16/18/31/33/35/39/45/51/52/56/58/59/66/68/73) were measured. Antibodies against HPV16/18 were higher after bivalent HPV vaccination (mean international units [IU] 1140.1 and 170.5 for HPV16 and 18, respectively) than after nonavalent vaccination (265.1 and 22.3 IUs, respectively). The bivalent vaccine commonly induced antibodies against the nonvaccine HPV types 31/33/35/45 or 58. The nonavalent vaccine induced higher antibodies against HPV6/11/31/33/45/52/58 and 35.


Subject(s)
Alphapapillomavirus , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Adult , Antibodies, Viral , Antibody Formation , Female , Human papillomavirus 16 , Human papillomavirus 18 , Humans , Papillomaviridae , Papillomavirus Infections/prevention & control , Vaccines, Combined
5.
J Infect Dis ; 224(1): 14-20, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33580261

ABSTRACT

BACKGROUND: Whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positivity among asymptomatic subjects reflects past or future disease may be difficult to ascertain. METHODS: We tested 9449 employees at Karolinska University Hospital, Stockholm, Sweden for SARS-CoV-2 RNA and antibodies, linked the results to sick leave records, and determined associations with past or future sick leave using multinomial logistic regression. RESULTS: Subjects with high amounts of SARS-CoV-2 virus, indicated by polymerase chain reaction (PCR) cycle threshold (Ct) value, had the highest risk for sick leave in the 2 weeks after testing (odds ratio [OR], 11.97; 95% confidence interval [CI], 6.29-22.80) whereas subjects with low amounts of virus had the highest risk for sick leave in the 3 weeks before testing (OR, 6.31; 95% CI, 4.38-9.08). Only 2.5% of employees were SARS-CoV-2 positive while 10.5% were positive by serology and 1.2% were positive in both tests. Serology-positive subjects were not at excess risk for future sick leave (OR, 1.06; 95% CI, .71-1.57). CONCLUSIONS: High amounts of SARS-CoV-2 virus, as determined using PCR Ct values, was associated with development of sickness in the next few weeks. Results support the concept that PCR Ct may be informative when testing for SARS-CoV-2. Clinical Trials Registration. NCT04411576.


Subject(s)
Asymptomatic Diseases , COVID-19/epidemiology , COVID-19/virology , Health Personnel , SARS-CoV-2 , Adult , Aged , Antibodies, Viral , COVID-19/diagnosis , Disease Progression , Female , Hospitals, University , Humans , Male , Mass Screening , Middle Aged , Polymerase Chain Reaction , RNA, Viral , SARS-CoV-2/genetics , Serologic Tests , Sick Leave/statistics & numerical data , Sweden/epidemiology , Young Adult
6.
Int J Cancer ; 149(12): 2083-2090, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34418082

ABSTRACT

The globally recommended public health policy for cervical screening is primary human papillomavirus (HPV) screening with cytology triaging of positives. To ensure optimal quality of laboratory services we have conducted regular audits of cervical smears taken before cervical cancer or cancer in situ (CIN3+) within an HPV-based screening program. The central cervical screening laboratory of Stockholm, Sweden, identified cases of CIN3+ who had had a previous cervical screening test up to 3 years before and randomly selected 300 cervical liquid-based cytology (LBC) samples for auditing. HPV testing with Roche Cobas was performed either at screening or with biobanked samples. HPV negative samples and subsequent biopsies were retrieved and tested with modified general primer HPV PCR and, if still HPV-negative, the LBCs and biopsies were whole genome sequenced. The Cobas 4800 detected HPV in 1020/1052 (97.0%) LBC samples taken before CIN3+. Further analyses found HPV in 28 samples, with nine of those containing HPV types not targeted by the Cobas 4800 test. There were 4 specimens (4/1052, 0.4%) where no HPV was detected. By comparison, the proportion of CIN3+ cases that were positive in a previous cytology were 91.6%. We find that the routine HPV screening test had a sensitivity in the real-life screening program of 97.0%. Regular laboratory audits of cervical samples taken before CIN3+ can be readily performed within a real-life screening program and provide assurance that the laboratory of the real-life program has the expected performance.


Subject(s)
Alphapapillomavirus/isolation & purification , Clinical Laboratory Services/statistics & numerical data , Mass Screening/statistics & numerical data , Papillomavirus Infections/diagnosis , Uterine Cervical Neoplasms/prevention & control , Adult , Aged , Cervix Uteri/pathology , Cervix Uteri/virology , Clinical Audit/statistics & numerical data , Clinical Laboratory Services/organization & administration , False Negative Reactions , Female , Humans , Mass Screening/methods , Mass Screening/standards , Middle Aged , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Sweden , Triage/methods , Triage/standards , Triage/statistics & numerical data , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaginal Smears/standards , Vaginal Smears/statistics & numerical data , Young Adult
7.
Am J Epidemiol ; 190(4): 506-514, 2021 04 06.
Article in English | MEDLINE | ID: mdl-32639531

ABSTRACT

The elimination of cervical cancer rests on high efficacy of human papillomavirus (HPV) vaccines. The HPV type distribution among cases of invasive cervical cancer (ICC) is used to make predictions about the impact of eliminating different types of HPV, but accumulating evidence of differences in age-specific cancer incidence by HPV type exists. We used one of the largest population-based series of HPV genotyping of ICCs (n = 2,850; Sweden, 2002-2011) to estimate age-specific ICC incidence by HPV type and obtain estimates of the cancer-protective impact of the removal of different HPV types. In the base case, the age-specific ICC incidence had 2 peaks, and the standardized lifetime risk (SLTR, the lifetime number of cases per birth cohort of 100,000 females) for HPV-positive ICC was 651 per 100,000 female births. In the absence of vaccine types HPV 16 and HPV 18, the SLTR for ICC was reduced to 157 per 100,000 female births (24% of HPV-positive SLTR). Elimination of all 9 types that can currently be vaccinated against reduced the remaining SLTR to 47 per 100,000 female births (7%), the remaining ICC incidence only slowly increasing with age. In conclusion, after elimination of vaccine-protected HPV types, very few cases of ICC will be left, especially among fertile, reproductive-age women.


Subject(s)
Papillomaviridae/immunology , Registries , Uterine Cervical Neoplasms/epidemiology , Vaccination/methods , Adult , Age Factors , Female , Follow-Up Studies , Humans , Incidence , Papillomavirus Infections , Retrospective Studies , Sweden/epidemiology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology
8.
Br J Cancer ; 123(12): 1790-1795, 2020 12.
Article in English | MEDLINE | ID: mdl-33020595

ABSTRACT

BACKGROUND: Human papillomavirus (HPV) is a necessary cause of cervical cancer, although some invasive cervical cancers may test negative by HPV PCR. We previously requested all invasive cervical cancers in Sweden during 10 years and subjected them to PCR. We also optimised methods for deep sequencing of formalin-fixed paraffin-embedded samples. METHODS: Using Novaseq 6000, we simultaneously sequenced total DNA and cDNA from 392 HPV PCR-negative cervical cancers. Non-human reads were queried against all known HPVs. The complete database now contains PCR and/or deep sequencing data on 2850 invasive cervical cancers. RESULTS: HPV sequences were detected in 169/392 of HPV PCR-negative cervical cancers. Overall, 30 different HPV types were detected, but only 5 types were present in proportions above 3% of cancers. More than 92% of tumours were HPV-positive in PCR and/or sequencing (95% confidence interval: 91.1-93.1%). Exploring possible reasons for failure to previously detect HPV suggest that more sensitive type-specific PCRs for HPV 31, 33, 45 and 73 targeting retained regions of HPV would have detected most of these (117/392). CONCLUSIONS: Unbiased deep sequencing provides comprehensive data on HPV types in cervical cancers and appears to be an important tool for quality assurance of HPV screening.


Subject(s)
Alphapapillomavirus/isolation & purification , High-Throughput Nucleotide Sequencing/methods , Uterine Cervical Neoplasms/virology , Adult , Aged , Aged, 80 and over , Confidence Intervals , DNA, Viral/isolation & purification , Female , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Middle Aged , Negative Results/statistics & numerical data , Reverse Transcriptase Polymerase Chain Reaction , Sweden , Uterine Cervical Neoplasms/pathology
9.
J Gen Virol ; 101(3): 265-270, 2020 03.
Article in English | MEDLINE | ID: mdl-31859615

ABSTRACT

Introduction. Cervical cancer is caused by human papillomavirus (HPV), but some cases may test HPV-negative. We previously tested 2850 Swedish cases and found that 394/2850 (13.8 %) cases tested HPV DNA-negative by PCR. Sequencing is the most thorough method to assess HPV status.Aim. We wished to assess whether deep sequencing might detect HPV sequences among these HPV-negative cervical cancer specimens, and to increase the likelihood of detecting transcriptionally active infections.Methodology. Out of the 2850 cancer cases, we sequenced a random sample of 92 HPV PCR-negative cervical cancers and 34 HPV PCR-positive cervical cancers. Four pools of blank blocks were sequenced as negative controls. To enrich for mRNA - a hallmark of active viral infection - the samples were extracted, reverse-transcribed, rRNA-depleted and then sequenced using the NovaSeq 6000 system (Illumina, USA). High-quality reads were aligned to the human genome and non-human reads were queried against HPV proteins.Results. We obtained a median of 23 million paired reads per sample. HPV was detected in 31/34 HPV PCR-positive cases. Among cases negative for HPV by PCR, 48/92 (52.2 %) contained HPV sequences, with HPV33 being the most commonly detected type among these (14/48 cases, 29.2 %). Comparison of the ratio of exon and intron sequences found that the sequenced material contained both DNA and RNA. Splice junctions were detected in 12 cases.Conclusion. Apparently, some cervical cancers contain HPV that is difficult to detect by PCR. Sequencing may be a helpful tool for additional quality assurance for HPV testing methods.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Negative Results , Papillomaviridae/genetics , Papillomavirus Infections/diagnosis , Polymerase Chain Reaction/methods , Uterine Cervical Neoplasms/virology , Adult , Aged , Aged, 80 and over , Cervix Uteri/pathology , Cervix Uteri/virology , DNA, Viral/genetics , Female , Genotype , Humans , Middle Aged , Papillomavirus Infections/virology , Sequence Analysis, DNA , Sweden
10.
BMC Cancer ; 20(1): 396, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32375689

ABSTRACT

BACKGROUND: To ensure the highest quality of human papillomavirus (HPV) testing in primary cervical cancer screening, novel HPV assays must be evaluated in accordance with the international guidelines. Furthermore, HPV assay with genotyping capabilities are becoming increasingly important in triage of HPV positive women in primary HPV screening. Here we evaluate a full genotyping HPV assay intended for primary screening. METHODS: The CLART® HPV4S (CLART4S) assay is a newly developed full-genotyping assay detecting 14 oncogenic (16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, 68) and two non-oncogenic HPV genotypes (6, 11). It was evaluated using SurePath and ThinPrep screening samples collected from the Danish and Swedish cervical cancer screening programs, respectively. For calculation of sensitivity, 81 SurePath and 80 ThinPrep samples with confirmed ≥CIN2 were assessed. For clinical specificity analysis, 1184 SurePath and 1169 ThinPrep samples from women with

Subject(s)
DNA, Viral/analysis , Early Detection of Cancer/methods , Papillomaviridae/genetics , Papillomavirus Infections/complications , Practice Guidelines as Topic/standards , Specimen Handling/methods , Uterine Cervical Neoplasms/diagnosis , Adult , Aged , DNA, Viral/genetics , Female , Follow-Up Studies , Genotype , Humans , Middle Aged , Papillomaviridae/isolation & purification , Papillomavirus Infections/virology , Prognosis , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Young Adult , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/virology
11.
Int J Cancer ; 145(3): 785-796, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30719706

ABSTRACT

Oncogenic non-vaccine human papillomavirus (HPV) types may conceivably fill the vacated ecological niche of the vaccine types. The likelihood of this may differ by the risk of acquiring HPV infections. We examined occurrence of HPV types among vaccinated and unvaccinated subgroups of 1992-1994 birth cohorts with differing acquisition risks up to 9 years post-implementation of HPV vaccination in 33 Finnish communities randomized to: Arm A (gender-neutral HPV16/18 vaccination), Arm B (girls-only HPV16/18 vaccination and hepatitis B-virus (HBV) vaccination of boys), and Arm C (gender-neutral HBV vaccination). Out of 1992-1994 born resident boys (31,117) and girls (30,139), 8,618 boys and 15,615 girls were vaccinated, respectively, with 20-30% and 50% coverage in 2007-2009. In 2010-2013, 8,868 HPV16/18 and non-HPV vaccinated females, and in 2014-2016, 5,574 originally or later (2010-2013) HPV16/18 vaccinated females attended two cervical sampling visits, aged 18.5 and 22-years. The samples were typed for HPV6/11/16/18/31/33/35/39/45/51/52/56/58/59/66/68 using PCR followed by MALDI-TOF MS. HPV prevalence ratios (PR) between Arms A/B vs. C were calculated for Chlamydia trachomatis positives (core-group), and negatives (general population minus core group). At both visits the vaccine-protected HPV type PRs did not significantly differ between the core-group and non-core group. Among the vaccinated 18-year-olds, HPV51 occurrence was overall somewhat increased (PRcore = 1.4, PRnon-core. = 1.4) whereas the HPV52 occurrence was increased in the core-group only (PRcore = 2.5, PRnon-core = 0.8). Among the non-HPV vaccinated 18-year-olds, the HPV51/52 PRs were higher in the core-group (PRcore = 3.8/1.8, PRnon-core = 1.2/1.1). The 22-year-olds yielded no corresponding observations. Monitoring of the sexual risk-taking core-group may detect early tendencies for HPV type replacement.


Subject(s)
Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Papillomavirus Infections/epidemiology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Risk-Taking , Sexual Behavior/statistics & numerical data , Adolescent , Adult , Female , Finland/epidemiology , Human papillomavirus 18/isolation & purification , Humans , Male , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Seroepidemiologic Studies , Sex Factors , Unsafe Sex , Young Adult
12.
PLoS Med ; 15(10): e1002666, 2018 10.
Article in English | MEDLINE | ID: mdl-30273338

ABSTRACT

BACKGROUND: High-risk human papillomavirus (hrHPV) infection is established as the major cause of invasive cervical cancer (ICC). However, whether hrHPV status in the tumor is associated with subsequent prognosis of ICC is controversial. We aim to evaluate the association between tumor hrHPV status and ICC prognosis using national registers and comprehensive human papillomavirus (HPV) genotyping. METHODS AND FINDINGS: In this nationwide population-based cohort study, we identified all ICC diagnosed in Sweden during the years 2002-2011 (4,254 confirmed cases), requested all archival formalin-fixed paraffin-embedded blocks, and performed HPV genotyping. Twenty out of 25 pathology biobanks agreed to the study, yielding a total of 2,845 confirmed cases with valid HPV results. Cases were prospectively followed up from date of cancer diagnosis to 31 December 2015, migration from Sweden, or death, whichever occurred first. The main exposure was tumor hrHPV status classified as hrHPV-positive and hrHPV-negative. The primary outcome was all-cause mortality by 31 December 2015. Five-year relative survival ratios (RSRs) were calculated, and excess hazard ratios (EHRs) with 95% confidence intervals (CIs) were estimated using Poisson regression, adjusting for education, time since cancer diagnosis, and clinical factors including age at cancer diagnosis and International Federation of Gynecology and Obstetrics (FIGO) stage. Of the 2,845 included cases, hrHPV was detected in 2,293 (80.6%), and we observed 1,131 (39.8%) deaths during an average of 6.2 years follow-up. The majority of ICC cases were diagnosed at age 30-59 years (57.5%) and classified as stage IB (40.7%). hrHPV positivity was significantly associated with screen-detected tumors, young age, high education level, and early stage at diagnosis (p < 0.001). The 5-year RSR compared to the general female population was 0.74 (95% CI 0.72-0.76) for hrHPV-positive cases and 0.54 (95% CI 0.50-0.59) for hrHPV-negative cases, yielding a crude EHR of 0.45 (95% CI 0.38-0.52) and an adjusted EHR of 0.61 (95% CI 0.52-0.71). Risk of all-cause mortality as measured by EHR was consistently and statistically significantly lower for cases with hrHPV-positive tumors for each age group above 29 years and each FIGO stage above IA. The difference in prognosis by hrHPV status was highly robust, regardless of the clinical, histological, and educational characteristics of the cases. The main limitation was that, except for education, we were not able to adjust for lifestyle factors or other unmeasured confounders. CONCLUSIONS: In this study, women with hrHPV-positive cervical tumors had a substantially better prognosis than women with hrHPV-negative tumors. hrHPV appears to be a biomarker for better prognosis in cervical cancer independent of age, FIGO stage, and histological type, extending information from already established prognostic factors. The underlying biological mechanisms relating lack of detectable tumor hrHPV to considerably worse prognosis are not known and should be further investigated.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/virology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/virology , Papillomaviridae/genetics , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/virology , Adenocarcinoma/pathology , Adult , Aged , Biosimilar Pharmaceuticals , Carcinoma, Squamous Cell/pathology , Female , Genotype , Humans , Middle Aged , Neoplasm Invasiveness , Papillomaviridae/isolation & purification , Prognosis , Prospective Studies , Registries , Survival Rate , Sweden/epidemiology , Uterine Cervical Neoplasms/pathology
13.
Br J Cancer ; 119(9): 1163-1168, 2018 10.
Article in English | MEDLINE | ID: mdl-30344308

ABSTRACT

BACKGROUND: HPV genomic variation may be involved in viral carcinogenesis. METHODS: In a national register-based nested case-control study, we retrieved archival smears from baseline cytologically normal women who later developed cancer in situ (CIS), squamous cervical cancer (SCC) or remained free of disease. These smears were previously HPV tested by PCR and HPV16 was the strongest risk factor. We now used the Illumina NextSeq platform to sequence HPV16 genomes in cervical smears from 242 women who later developed CIS/CIN3 (n = 134), SCC (n = 92) or remained healthy (n = 16). RESULTS: The median sequence depth per sample was high (11,288×). For 218/242 samples (>90%), we covered ≥80% of the complete HPV16 genome with sequencing median depths of >200×. We identified a wide range of unique isolates and 147 novel SNPs across the 218 samples. Most women (97%) had HPV16 lineage A infection, with the sublineages being A1 (66.1%), A2 (28.9%) and A4 (1.8%), respectively. The least variable gene was the E7 (3.4% variability), where 170/204 case women (83%) displayed a fully conserved sequence. There were no obvious differences by disease outcome (CIS or SCC). CONCLUSIONS: We found a high number of novel SNPs. The E7 gene was hypovariable both among women later developing CIN3/CIS, and SCC, respectively.


Subject(s)
Carcinoma in Situ/virology , Genome, Viral , Human papillomavirus 16/genetics , Neoplasm Invasiveness , Uterine Cervical Neoplasms/virology , Adolescent , Adult , Carcinoma in Situ/diagnosis , Carcinoma in Situ/epidemiology , Case-Control Studies , Female , Humans , INDEL Mutation , Middle Aged , Polymorphism, Single Nucleotide , Prospective Studies , Registries , Sweden/epidemiology , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/epidemiology , Vaginal Smears , Young Adult
14.
Br J Cancer ; 118(10): 1377-1381, 2018 05.
Article in English | MEDLINE | ID: mdl-29559733

ABSTRACT

BACKGROUND: The Swedish National Cervical Screening Registry collects and evaluates comprehensive, nationwide health data to optimise organised cervical cancer prevention. Since all cervical cancer specimens are saved in biobanks, population-based data from the specimens should be available for analysis and linkage with other health information. METHODS: We identified all cervical cancers diagnosed in Sweden during 2002-2011 (4254 confirmed cases) and requested the tissue blocks to retrieve human papillomavirus (HPV) genotype data using general primer PCR with Luminex genotyping and real-time PCR targeting the E6/E7 regions of HPV16/18. RESULTS: We obtained blocks from 2932/4254 (69%) of cases. Valid HPV genotyping data was retrieved for 2850 cases (97%). The most common type was HPV16 (60%), followed by HPV18 (19%), HPV45 (7%), HPV31 (3%), HPV33 (2%), HPV52 (2%), HPV39 (1%), HPV70 (1%), HPV56 (1%), HPV35 (1%), HPV58 (1%) and HPV59 (1%). Ninety-six percent of all HPV-positive cases had a single infection. Eighty-nine cases were HPV-positive only when testing for the HPV16/18-E6/E7 region. CONCLUSIONS: We present one of the largest series of HPV-genotyped cervical cancers to date. The systematic collection of cervical cancer HPV genotyping data by the screening registry will facilitate prevention and monitoring of HPV type-specific disease burden.


Subject(s)
Early Detection of Cancer , Papillomaviridae/genetics , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/virology , DNA, Viral/genetics , DNA, Viral/isolation & purification , Female , Genotype , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/classification , Papillomaviridae/pathogenicity , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/classification , Papillomavirus Infections/genetics , Repressor Proteins/genetics , Sweden/epidemiology , Uterine Cervical Neoplasms/classification , Uterine Cervical Neoplasms/genetics
15.
Int J Cancer ; 141(12): 2498-2504, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28840939

ABSTRACT

Most cancer forms known to be caused by viruses are increased among the immunosuppressed, but several cancer forms without established viral etiology are also increased, notably nonmelanoma skin carcinoma (NMSC). We followed all 13,429 solid organ transplantation patients in Sweden for cancer occurrence after transplantation. We requested these tumor specimens and sequenced the first 89 specimens received (62 NMSCs, 27 other cancers). The sequences were analyzed for viruses based on two bioinformatics algorithms (paracel-blast (sensitive for detection of known viruses) and hidden Markov model (HMM; sensitive for distantly related viruses)). Among the 62 NMSCs, the virus family detected in the largest proportion of specimens was Mimiviridae (9/62 NMSCs). The majority of the virus-related reads belonged to Papillomaviridae. The HMM analysis identified 86 additional previously not described viral contigs related to 11 virus families, with reads related to Mimiviridae being the most common (detected in 28/62 NMSCs) with the most prevalent contig (Mimivirus SE906, 1937 bp) detected in 17/62 NMSCs. Among the 27 other cancers, viral sequences were detected in only 5 specimens by blast analysis, compared to in all 27 specimens by HMM (Mimiviridae, Poxviridae, Phycodnaviridae and virus-related sequences yet unclassified to any family). 99% of the virus reads belonged to a single previously not described sequence (Mimivirus SE996, 911 bp). A multitude of viruses is readily detectable in specimens with cancers occurring among the immunosuppressed, with sequences related to Mimiviridae being the most prevalent. Further research would be needed to elucidate the biological significance of the viruses.


Subject(s)
Neoplasms/immunology , Organ Transplantation/adverse effects , Sequence Analysis, DNA/methods , Virus Diseases/epidemiology , Viruses/classification , Algorithms , Computational Biology/methods , Humans , Immunocompromised Host , Markov Chains , Mimiviridae/genetics , Mimiviridae/isolation & purification , Neoplasms/virology , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Sweden , Viruses/genetics
16.
Int J Cancer ; 136(11): 2546-55, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25388227

ABSTRACT

Non-melanoma skin cancers commonly contain Human Papillomavirus (HPV), but the types found have varied depending on the polymerase chain reaction (PCR) primer systems used. Whole genome amplified DNA (not amplified by any specific PCR primers) from 91 skin lesions [41 squamous cell skin carcinomas (SCCs), 8 keratoacanthomas, 22 actinic keratoses, 3 basal cell carcinomas and 17 SCCs in situ] were sequenced. All samples were sequenced both at 160 Mb and 1.8 Gb sequencing depth per sample. The sequences from 10 different HPVs in 47/91 specimens were found. Sequences represented four established HPV types (HPV types 16, 22, 120, 124), two previously known putative types (present in GenBank) and four previously unknown HPV sequences (new putative types). The most commonly detected virus was cloned, sequenced and designated as HPV197. Type-specific real-time PCR detected HPV197 in 34/91 specimens. For comparison, a pool of the same samples after general primer PCR amplification was also sequenced. This revealed 40 different HPVs, but only two HPV types were detected both with sequencing without prior PCR and with sequencing PCR amplicons, suggesting that sequencing without prior PCR gives a more unbiased representation of the HPVs present. In summary, it was found that HPV can be sequenced from most skin disease specimens and HPV197 appeared to be the most commonly present virus.


Subject(s)
Papillomaviridae/classification , Papillomaviridae/isolation & purification , Skin Neoplasms/virology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/virology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cloning, Molecular , DNA, Viral/genetics , Humans , Keratoacanthoma/genetics , Keratoacanthoma/virology , Keratosis, Actinic/genetics , Keratosis, Actinic/virology , Molecular Sequence Data , Papillomaviridae/genetics , Sequence Analysis, DNA , Skin Neoplasms/genetics , Skin Neoplasms/pathology
17.
PLoS One ; 19(7): e0305285, 2024.
Article in English | MEDLINE | ID: mdl-38990856

ABSTRACT

INTRODUCTION: Serological surveys of the prevalence of SARS-CoV-2 are instrumental to understanding the course of the COVID-19 epidemic. We evaluate the seroprevalence of SARS-CoV-2 among young adult Finnish females residing in 25 communities all over Finland from 2020 until 2022. METHODS: Between 1st March 2020 and 30th June 2022, 3589 blood samples were collected from 3583 women born in 1992-95 when aged 25 or 28 years old attending the follow-up of an ongoing population-based trial of cervical screening strategies. The crude and population standardized SARS-CoV-2 seroprevalence was measured using nucleocapsid (induced by infection) and spike wild-type (WT) protein (induced both by infection and by vaccination) antigens over time and stratified by place of residence (inside or outside the Helsinki metropolitan region). RESULTS: During 2020 (before vaccinations), spike-WT and nucleocapsid IgG antibodies followed each other closely, at very low levels (<5%). Spike-WT seropositivity increased rapidly concomitant with mass vaccinations in 2021 and reached 96.3% in the 2nd quartile of 2022. Antibodies to nucleocapsid IgG remained relatively infrequent throughput 2020-2021, increasing rapidly in the 1st and 2nd quartiles of 2022 (to 19.7% and 56.6% respectively). The nucleocapsid IgG seropositivity increased more profoundly in participants residing in the Helsinki metropolitan region (4.5%, 8.4% and 43.9% in 2020, 2021 and 2022 respectively) compared to those residing in communities outside the capital region (4.5%, 4.3% and 34.7%). CONCLUSIONS: Low SARS-CoV-2 infection-related seroprevalence during 2020-2021 suggest a comparatively successful infection control. Antibodies to the SARS-CoV-2 WT spike protein became extremely common among young women by the end of 2021, in line with the high uptake of SARS-CoV-2 vaccination. Finally, the rapid increase of seroprevalences to the SARS-CoV-2 nucleocapsid protein during the first and second quartile of 2022, imply a high incidence of infections with SARS-CoV-2 variants able to escape vaccine-induced protection.


Subject(s)
Antibodies, Viral , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Female , Finland/epidemiology , COVID-19/epidemiology , COVID-19/immunology , Adult , Seroepidemiologic Studies , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Antibodies, Viral/blood , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Coronavirus Nucleocapsid Proteins/immunology
18.
Nat Commun ; 15(1): 3679, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693149

ABSTRACT

HPV vaccination with concomitant HPV-based screening of young women has been proposed for faster cervical cancer elimination. We describe the baseline results of a population-based trial of this strategy to reduce the incidence of HPV. All 89,547 women born 1994-1999 and resident in the capital region of Sweden were personally invited to concomitant HPV vaccination and HPV screening with 26,125 women (29.2%) enrolled between 2021-05-03 and 2022-12-31. Baseline HPV genotyping of cervical samples from the study participants finds, compared to pre-vaccination prevalences, a strong decline of HPV16 and 18 in birth cohorts previously offered vaccination, some decline for cross-protected HPV types but no decline for HPV types not targeted by vaccines. Our dynamic transmission modelling predicts that the trial could reduce the incidence of high-risk HPV infections among the 1994-1998 cohorts by 62-64% in 3 years. Baseline results are prevalences of HPV infection, validated transmission model projections, and power estimates for evaluating HPV incidence reductions at follow-up (+/-0.1% with 99.9% confidence). In conclusion, concomitant HPV vaccination and HPV screening appears to be a realistic option for faster cervical cancer elimination. Clinicaltrials.gov identifier: NCT04910802; EudraCT number: 2020-001169-34.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/epidemiology , Papillomavirus Infections/epidemiology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/therapeutic use , Adult , Sweden/epidemiology , Young Adult , Vaccination , Adolescent , Incidence , Mass Screening , Prevalence , Middle Aged , Early Detection of Cancer , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Human papillomavirus 18/genetics , Human papillomavirus 18/immunology , Human Papillomavirus Viruses
19.
J Clin Virol ; 171: 105657, 2024 04.
Article in English | MEDLINE | ID: mdl-38401369

ABSTRACT

BACKGROUND: Some high-grade cervical lesions and cervical cancers (HSIL+) test negative for human papillomavirus (HPV). The HPV-negative fraction varies between 0.03 % and 15 % between different laboratories. Monitoring and extended re-analysis of HPV-negative HSIL+ could thus be helpful to monitor performance of HPV testing services. We aimed to a) provide a real-life example of a quality assurance (QA) program based on re-analysis of HPV-negative HSIL+ and b) develop international guidance for QA of HPV testing services based on standardized identification of apparently HPV-negative HSIL+ and extended re-analysis, either by the primary laboratory or by a national HPV reference laboratory (NRL). METHODS: There were 116 initially HPV-negative cervical specimens (31 histopathology specimens and 85 liquid-based cytology samples) sent to the Swedish HPV Reference Laboratory for re-testing. Based on the results, an international QA guidance was developed through an iterative consensus process. RESULT: Standard PCR testing detected HPV in 55.2 % (64/116) of initially "HPV-negative" samples. Whole genome sequencing of PCR-negative samples identified HPV in an additional 7 samples (overall 61.2 % HPV positivity). Reasons for failure to detect HPV in an HSIL+ lesion are listed and guidance to identify cases for extended re-testing, including which information should be included when referring samples to an NRL are presented. CONCLUSION: Monitoring the proportion of and reasons for failure to detect HPV in HSIL+ will help support high performance and quality improvement of HPV testing services. We encourage implementation of QA strategies based on re-analysis of "HPV negative" HSIL+ samples.


Subject(s)
Papillomavirus Infections , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Dysplasia/diagnosis , Human Papillomavirus Viruses , Papillomavirus Infections/diagnosis , Mass Screening/methods , Papillomaviridae/genetics
20.
Obstet Gynecol ; 142(3): 679-687, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37535949

ABSTRACT

OBJECTIVE: To evaluate the risk for cervical intraepithelial neoplasia grade 3 (CIN 3) or worse (including adenocarcinoma in situ [AIS] and invasive cervical cancer) associated with non-16/18 human papillomavirus (HPV) types (other HPV) among women with atypical glandular cells (AGC) in cervical cytology. METHODS: This population-based cohort study evaluates the risk of CIN 3 or worse associated with other HPV types. Human papillomavirus genotyping was performed on Pap tests collected in Sweden from 341 women with AGC that were positive for other HPV types from February 17, 2014, to December 31, 2018. The women were followed for histopathologic outcomes using comprehensive registry linkages until December 31, 2019. Cumulative incidence proportions of CIN 3 or worse by specific HPV type were calculated using 1-minus Kaplan-Meier function. Hazard ratios (HRs) for CIN 3 or worse were generated using multivariate Cox regression. RESULTS: Of 341 women, 134 (39.3%) had CIN 3-AIS, but there were only five (1.5%) women in the cohort with invasive cervical cancer. Human papillomavirus 45 preceded 80.0% of invasive cervical cancer cases. Among women positive for HPV33, 82.9% (95% CI 58.0-97.3%) had CIN 3 or worse during follow-up. Positivity for HPV31 conferred the highest HR for CIN 3 or worse relative to other types, both in primary cytology and primary HPV screening (HR 2.71, 95% CI 1.47-5.00 and HR 3.41, 95% CI 1.95-5.96, respectively). CONCLUSION: Among non-16/18 HPV types in AGC, HPV31 and 33 had the highest risk for CIN 3 or worse, whereas most of the women with invasive cancer were positive for HPV45. Extended HPV genotyping may be helpful for the management of AGC.


Subject(s)
Papillomavirus Infections , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Female , Humans , Male , Uterine Cervical Neoplasms/pathology , Human Papillomavirus Viruses , Cohort Studies , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Uterine Cervical Dysplasia/diagnosis , Vaginal Smears , Papillomaviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL