Next Article in Journal
Female Sex but Not Oestrogen Receptor Expression Predicts Survival in Advanced Gastroesophageal Adenocarcinoma—A Post-hoc Analysis of the GO2 Trial
Previous Article in Journal
Inhibition of PLK1 Destabilizes EGFR and Sensitizes EGFR-Mutated Lung Cancer Cells to Small Molecule Inhibitor Osimertinib
Previous Article in Special Issue
Current Treatment Practices and Prognostic Factors in Early-Stage Ovarian Cancer—An Analysis of the NOGGO/JAGO
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Circulating HPV DNA as a Biomarker for Pre-Invasive and Early Invasive Cervical Cancer: A Feasibility Study

1
UCL Elizabeth Garrett Anderson Institute for Women’s Health, Faculty of Population Health Sciences, University College London, Medical School Building, 74 Huntley Street, London WC1E 6AU, UK
2
The Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
3
The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
4
Translational Research Lab, Department of Women’s Cancer, IfWH, Ground Floor POGB, 72 Huntley Street, London WC1E 6DD, UK
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(9), 2590; https://doi.org/10.3390/cancers15092590
Submission received: 18 February 2023 / Revised: 29 March 2023 / Accepted: 24 April 2023 / Published: 2 May 2023

Abstract

:

Simple Summary

Over 95% of cervical cancers are caused by the human papilloma virus (HPV). Locally advanced HPV-related cancers release tiny bits of tumour containing HPV-DNA into the blood (cHPV-DNA). The presence of cHPV-DNA in the blood can serve as a potential marker of cancer and should be absent in pre-cancerous conditions (cervical intraepithelial neoplasia (CIN). We have developed an ultra-sensitive and specific test to measure blood cHPV-DNA levels. We performed a feasibility study to confirm our expectation that cHPV-DNA is not found in the blood of patients with CIN but is found in the blood of patients with very early cancers. In this study, we have confirmed that cHPV-DNA is absent in the plasma of women with CIN. In early cervical tumours, there was a low detection rate of cHPV-DNA. Therefore, more sensitive tests are required before cHPV-DNA can be used for the detection of very early cervical cancers.

Abstract

Background: High-risk HPV infection is responsible for >99% of cervix cancers (CC). In persistent infections that lead to cancer, the tumour breaches the basement membrane, releasing HPV-DNA into the bloodstream (cHPV-DNA). A next-generation sequencing assay (NGS) for detection of plasma HPV circulating DNA (cHPV-DNA) has demonstrated high sensitivity and specificity in patients with locally advanced cervix cancers. We hypothesised that cHPV-DNA is detectable in early invasive cervical cancers but not in pre-invasive lesions (CIN). Methods: Blood samples were collected from patients with CIN (n = 52) and FIGO stage 1A-1B CC (n = 12) prior to treatment and at follow-up. DNA extraction from plasma, followed by NGS, was used for the detection of cHPV-DNA. Results: None of the patients with pre-invasive lesions were positive for CHPV-DNA. In invasive tumours, plasma from one patient (10%) reached the threshold of positivity for cHPV-DNA in plasma. Conclusion: Low detection of cHPV-DNA in early CC may be explained by small tumour size, poorer access to lymphatics and circulation, and therefore little shedding of cHPV-DNA in plasma at detectable levels. The detection rate of cHPV-DNA in patients with early invasive cervix cancer using even the most sensitive of currently available technologies lacks adequate sensitivity for clinical utility.

1. Background

In the UK, cervical cancer is the 14th most common cancer in females [1]. Overall five-year survival is approximately 61% but is dependent on the stage at diagnosis, with 96% of women surviving the disease for a year or more if diagnosed early, versus 50% who are diagnosed at a later stage [1]. Equally, age at diagnosis plays a role, with around 90% of women diagnosed between ages 15 and 39 surviving for 5 years or more, compared to 25% of those aged 80 or over.
Risk factors for cervical cancer are multiple [2], including smoking [3], sexual practices [4], and exposure to and persistence of high-risk human papillomavirus (HRHPV)—which is detected in 99.7% of cervical cancers and has consequently been listed as a carcinogen by the International Agency for Research on Cancer (IARC) [1,5].
Many cancers are known to shed fragments of DNA (circulating tumour DNA, ctDNA) into the bloodstream. Most cervical cancers are causally related to the human papilloma virus (HPV) [6]. In the tumour, HPV DNA is integrated into the host genome or present in episomal form [7] and can be released into the bloodstream. Thus, circulating HPV-DNA (cHPV-DNA) can potentially be used as a detection marker for HPV-related cervical cancer.
Various studies investigating cHPV-DNA as a tumour marker in cervical cancer patients have demonstrated between 11% and 45% positivity rates of cHPV-DNA in patients with invasive cervical cancer at diagnosis, suggesting a low sensitivity of the PCR assays [8,9,10].
cHPV-DNA has also been detected in HPV-positive head and neck cancers, with many of the studies employing PCR methods, which have a sensitivity of 19–79% in locally advanced diseases. The use of digital droplet PCR (ddPCR) using probes for HPV 16 and 33 has been shown to increase the detection of HPV ctDNA in oropharyngeal squamous cell carcinomas (OPSCC) to almost 96% [11]. Similarly, improved detection using ddPCR has been described in cervical cancer in the metastatic setting [12]. There has been limited success in detecting cHPV-DNA for cervical cancer diagnosis due to the lack of sensitivity of conventional PCR assays [13].
Traditionally, HPV genotyping involved the amplification of a region of the L1 gene with next-generation sequencing (NGS) using specific primers. Lee et al. developed an amplicon-based next-generation sequencing assay to detect circulating plasma viral DNA (‘HPV16-detect’) to assess the response to chemoradiotherapy in locally advanced head and neck cancer [14]. This novel NGS assay comprises a 39-amplicon single pool panel that covers 34 distinct regions of the HPV 16 genome, as well as five amplicons of human reference genes. In their cohort, the assay demonstrated 100% sensitivity and 93% specificity in the detection of HPV 16-positive head and neck cancers at diagnosis. In addition, by tracking HPV through and post-chemoradiation, they were able to predict response and residual disease, demonstrating that cHPV-DNA can be used as a marker of response to treatment.
In their other study, which included a cohort of HPV-related anal squamous cell carcinoma patients post-chemoradiotherapy, Lee et al. expanded the novel NGS assay to include HPV subtypes 16, 18, 31, 33, 35, 45, 52, and 58 (panHPV-detect) [15]. This included two primer pools covering distinct regions (single nucleotide polymorphisms) of the HRHPV genome to detect cHPV-DNA. In this study, they demonstrated 100% sensitivity and specificity of panHPV-detect in diagnosing the high-risk HPV subtypes prior to treatment of anal squamous cell carcinoma. Furthermore, panHPV-detect was able to identify those patients who either had residual disease post-treatment or relapsed a few months later [15].
There has been some work surrounding early detection and follow-up of treated cervical disease, with circulating DNA playing a promising role. However, these studies tend to include advanced cancers rather than early invasive cancers. In this prospective cohort study, we aimed to investigate the use of this highly sensitive assay—panHPV-detect—for the detection of cHPV-DNA in both pre-invasive cervical lesions and early invasive cervical cancer. As HPV does not enter the bloodstream in its infective phase, we hypothesized that cHPV-DNA would not be detected in pre-invasive cervical disease but would be detectable in early-stage cancers.

2. Methods

This study was approved by the institutional board (Ref. no. CCR 4157) and ethics committee (Ref. no. 14/NE/1055). Patients who were referred to the colposcopy clinic at UCLH with cervical smears showing high-grade dyskaryosis or persistent mild dyskaryosis and HPV-positives, and who were attending for cervical treatment in the form of Large Loop Excision of the Transformation Zone (LLETZ) or knife cone biopsy, were recruited. Inclusion criteria were those over the age of 18, referred with HSIL and HPV+ve, and able to give informed consent. The exclusion criteria were those with a previous diagnosis of cancer, previous CIN treatment, and other HPV-associated lesions (VIN/VaIN/AIN). Following written, informed consent, 20 mL of venous blood was collected in EDTA tubes prior to treatment and then at the next appointment following treatment to assess for cHPV-DNA. The histological findings were correlated with the panHPV-detect result. Women who could not tolerate treatment in the outpatient setting or were undergoing general anesthesia for treatment of a larger lesion were seen in the clinic at the time of consent for their surgery and recruited to this study. A venous blood sample was then taken on the morning of surgery.
A second cohort of patients with biopsy-proven early-stage (FIGO 2018 1A/1B) invasive disease was recruited from the UCLH Gynaecological Oncology clinic., in London. These patients were recruited to this study in the outpatient clinic based on radiological staging and prior to definitive treatment, which was typically a simple or radical hysterectomy.

2.1. Sample Size Calculation

In the head and neck cancer pilot study, HPV16-detect demonstrated 100% sensitivity and 93% specificity in detecting HPV16-positive cancers at diagnosis. To have 80% power to detect a true sensitivity of 99%, we calculated that at least 19 HPV-positive, invasive cancer patients needed to be recruited. As approximately 95% of cervical cancer patients are HPV-positive, we aimed to recruit 22 patients with early invasive carcinoma. To confirm 93% specificity for pre-invasive disease, we required 37 patients with high-grade squamous intraepithelial lesions (HSIL) to be recruited.

2.2. Sample Collection and Processing

Whole blood sampling and plasma extraction were conducted at the Translational Research Laboratory at UCL, London, UK. The frozen plasma samples were then transported to the Chester Beatty Lab at the Institute of Cancer Research (ICR), London, UK for further analysis.
Additionally, 20 mL of blood was centrifuged at 1600× g for 10 min within 48 h of collection. The resulting plasma was then centrifuged again at 1600× g for 10 min to remove any cell sediment, aliquoted, and frozen at −80 °C. DNA was extracted from 5 mL of plasma using the QIAamp Circulating Nucleic Acid Kit (Qiagen) according to the manufacturer’s instructions. DNA was eluted in 50 μL of AVE buffer and stored at −20 °C. Plasma DNA was quantified using a Bio-Rad QX200 ddPCR system (Bio-Rad, Hertfordshire, UK), using ribonuclease P (RNase P) as a reference gene, as previously described [16].
We performed an initial validation of panHPV-detect by testing its ability to correctly identify the HPV subtype in cervical cancer tissue samples. We obtained tumour DNA extracted from cervical cancer tissue previously typed for the HPV subtype using polymerase chain reaction (RT-PCR) for E6 and E7 mRNA using validated assays. Five samples for each of the eight high-risk HPV subtypes were obtained from the Scottish HPV archive. Samples from five HPV-negative patients with head and neck cancer were recruited in the head and neck cancer cohort of this study, and 19 patients with breast cancer enrolled in the prospective sample collection study (PlasmaDNA, CCR3297, REC Ref No: 10/H0805/50) were used as negative controls. HPV status was confirmed as negative with E7 RT-PCR performed on RNA extracted from the samples. None of the negative controls were known to have pre-cancerous lesions. Written informed consent was obtained from all participants. Following the initial validation, we investigated whether panHPV-detect was able to detect cHPV-DNA in the pre-treatment plasma of patients recruited in this study.
To classify HPV+ and HPV samples using panHPV-detect in tissue, we set a threshold whereby a sample was classified positive if there were 10 reads present from more than 30% of different HPV amplicons for each subtype. To assess the threshold for the number of amplicons needed for a positive panHPV-detect readout in plasma, a ROC analysis was used. In the first step, HPV status and subtype were assigned in tissue using E7 mRNA to separate the two groups. The number of amplicons with greater than 10 reads at baseline was inputted for each patient to find a suitable threshold for this parameter. Sorting the values in both HPV+ and negative groups and averaging adjacent values in the sorted list generated a list of thresholds. Based on the ROC analysis, a threshold that gave the greatest sensitivity and specificity for each HPV subtype was selected as the threshold for the classification of plasma as HPV DNA-positive. Based on the ROC analysis, a threshold of 7 amplicons with more than 10 reads were the thresholds that gave the greatest sensitivity and specificity and were selected as the thresholds for the classification of plasma as HPV DNA-positive.
DNA library preparation was performed, and then Ion Torrent qPCR Quantitation with final sequencing was performed at the Tumour Profiling Unit of the ICR. Ion Torrent Libraries were prepared using Ion Ampliseq Library Preparation kit 2.0 (Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer’s instructions using 5 ng of tissue DNA or 3 ng of plasma DNA per primer pool, except the volumes of the Ion Ampliseq Library Preparation kit and the Custom Primer Pools have been reduced by half. Reads were aligned to an amalgamated reference containing scaffolds for each of the HPV genotypes as well as the reference human targeting genes using default settings in module map4 of the TMAP on the Ion Torrent machine. Bedtools v2.23.0 (Salt Lake City, UT, USA) [17] was used to extract on-target reads from the aligned files with a minimum overlap of 90% with amplicons in the panel. Additionally, reads with a mapping quality of <30 were removed using Samtools v1.2 (Hinxton, UK) [18]. Reads were split into those covering human and HPV amplicons, and coverage of each portion and each genotype was calculated individually.
In order to classify HPV+ and HPV samples using panHPV-detect in tissue, we set a threshold whereby a sample was classified positive if there were 10 reads present from more than 7 different HPV amplicons for each subtype.

2.3. Cost of Sample Analysis

The cost of sample analysis was £132 per sample (Table S1, Figures S1 and S2). The bulk cost of the sample analysis is apportioned to the sequencing costs and the library preparation kit costs. We have optimized the library preparation protocol to use half the recommended volume of reagents for library preparation.

3. Results

We recruited 52 patients between November 2018 and January 2020. The COVID-19 pandemic necessitated the suspension of study recruitment and sample collection, including follow-up of already recruited patients, in January 2020. All non-urgent clinical studies were placed on hold as laboratory services and resources were diverted to the pandemic. In addition, there was a cessation of the cervical screening programme for 2 months, a restriction on colposcopy appointments, and the introduction of social distancing rules, which meant that further recruitment from and follow-up at clinics were halted.
None of the recruited patients had a history of prior HPV vaccination. Patient demographics can be found in Table 1.

3.1. Detection of CHPV-DNA in Baseline Samples

Of the 52 patients recruited, 12 had early invasive cervical cancer and 40 had pre-invasive disease (Table 2). The mean ages of participants with cancer and pre-invasive disease were 39 and 35 years, respectively. At the time of baseline blood collection, the final histology was not known; therefore, all samples were included for analysis. Analysis was performed blind to the referral HPV status. An adequate quantity of DNA could be extracted from 50 of the 52 patients, with two samples (9 and 11) failing the library preparation stage due to insufficient volume of DNA despite PCR amplification.
Using the pre-set threshold of 10 reads present from >7 HPV amplicons, only 2 of the 50 baseline samples were HPV positive. HPV reads were detected in 15 other samples but in no more than 6 amplicons per HPV subtype and therefore did not reach the threshold for positivity (Table 3).

3.2. Detection of cHPV-DNA in Early Invasive Cervical Cancer

Final histology was available after all plasma samples were analysed and DNA sequencing performed (including follow-up samples). Out of the 12 cancer samples, 2 were found to be at a more advanced stage (FIGO 2B) than the inclusion criteria and therefore had to be excluded from this study (1 of these reached the threshold for positivity for HPV). Of the remaining 10 samples, only one was positive for HPV as per threshold criteria. This sample came from a patient who had stage 1A1 cervical cancer and was found to be HPV 18-positive, which correlated with the HPV type on referral cytology. This patient was an ex-smoker, under 30 years old, and had a small tumour. The other five patients whose histology indicated FIGO stage 1A1 tumours, did not reach the threshold for HPV positivity in plasma. None of the four 1B1 tumours had HPV in the plasma at levels that reached the threshold for positivity.

3.3. Detection of cHPV-DNA in Pre-Invasive Cervical Disease

None of the plasma samples from patients with final histology showing CIN 1–3 or cervical glandular intraepithelial neoplasia (CGIN) reached the threshold for positivity for HPV (Table 3). In 10 of these samples, HPV amplicons were detected, but at numbers far less than the threshold. Combining the results from the detection of CHPV-DNA in pre-invasive and invasive disease gives a sensitivity of 10% (95% CI, 0.24–44.5%) and a specificity of 100% (95% CI, 91.2–100%). There was no correlation between the number of amplicons detected and the grade of CIN or stage of cancer.

3.4. Detection of cHPV-DNA in Follow-Up Samples

Only eighteen patients out of the cohort of 50 were followed up prior to the COVID-19 pandemic. Two of the patients (9 and 11) had to have follow-up samples analysed despite baseline samples not being available (all samples were blinded and run on the same chip, regardless of follow-up or baseline). It was not known at the time of the blood draw that the baseline plasma had failed the PCR and library preparation stages. Of these 18 follow-up samples, 10 had HPV reads detected but none at >6 amplicons, and therefore none of the follow-up samples reached thresholds for positivity (Table 4). Sample number 25, which was the only positive baseline sample—detecting HPV 18 prior to treatment—was negative at follow-up.
Of the 10 samples that had HPV reads at follow-up, only 2 correlated with the baseline HPV read—in one case, there was no baseline sample to compare. Seven samples detected HPV types that were either new or did not correlate with the baseline HPV types (Table 4).

4. Discussion

The aim of this project was to determine whether panHPV-detect could detect cHPV-DNA in the plasma of patients with early-stage cervical cancers, as it has previously been shown to have high sensitivity and specificity for locally advanced HPV-associated cervical, head and neck, and anal cancers [14,15]. Using the thresholds of positivity for head and neck and anal cancers, this study has demonstrated that none of the patients with pre-invasive disease had cHPV-DNA in their plasma (specificity 100%), which is compatible with the first of our study hypotheses: that pre-invasive disease does not shed detectable levels of cHPV-DNA into the circulation. However, we demonstrated an assay sensitivity of only 10% for early-stage invasive disease.
The low sensitivity of this study correlates with other similar studies. Pornthanakasem et al. investigated the diagnostic and prognostic potential of circulating cHPV-DNA in patients with cervical cancer [9]. They collected tissue from 63 patients with cervical cancer and obtained blood samples from the same cohort and from 20 healthy blood donors. Only 6/50 patients with HPV-associated cervical cancer had circulating DNA, indicating low sensitivity. However, circulating DNA was not found to be present in normal controls or in HPV-negative cervical cancer, indicating a high specificity. Furthermore, the HPV genomes from both the tumour and the plasma matched, indicating that the circulating viral DNA was derived from the tumour cells. This study also reported that HPV in the plasma was associated with distant metastases, recurrence within 1 year, and therefore a poor prognosis [9].
Sathish et al. also looked at cHPV-DNA in cervical cancer patients. They collected cervical biopsies and plasma samples from 58 women with invasive cervical cancer. In addition, they also collected samples from 10 women with the pre-invasive disease and 30 control women matched for age. All paired samples for the control group were negative for HPV DNA; however, only eight (11.8%) were positive for cHPV-DNA in the cervical cancer group. In the CIN group, all 10 patients were positive for HPV in cervical biopsies, but none were positive in plasma samples. Of the eight patients who were plasma DNA positive, seven were HPV 16-positive, and six were at stage 3–4 cancer. Cervical tissue and plasma pairs were correlated on sequencing [10].
Both the aforementioned studies used more conventional PCR techniques rather than the novel NGS assay used in this study, which we had expected to achieve greater sensitivity for the detection of disease.
A systematic review by Trigg et al. investigated the possible factors that influence the quality and yield of circulating free DNA [19]. The factors evaluated included the specimen type, collection tube type, time to processing, centrifugation protocols, and methods of cfDNA isolation and quantification. Based on the results of this systematic review, our protocols followed the recommendations suggested to maximise the yield of DNA from plasma. Of the two samples with insufficient DNA, the extractions were repeated but gave the same results, suggesting a true deficit in cfDNA in those samples rather than an error in technique.
We have carried out a similar study [20], which evaluated panHPV-detect in locally advanced cervical cancers in patients undergoing chemoradiotherapy. This study has demonstrated high sensitivity (88%) and specificity (100%) for our NGS assay in detecting cHPV-DNA at baseline and following treatment completion. This suggests that the stage or volume of tumour plays a role in the amount of circulating tumour DNA that may be released into the plasma.
Gu et al. published a recent meta-analysis of cHPV-DNA as a biomarker for cervical cancer [21]. They found 10 studies published between 2001 and 2018 that met their eligibility criteria and covered 684 patients with cervical cancer. Patients with primary or metastatic cervical cancers at stages 1–4 who were receiving all modalities of treatment were included. Tumour types were both squamous and adenocarcinomas, with a greater proportion being squamous cell tumours. None of the control or pre-invasive disease groups had positive cHPV-DNA in the serum. The overall pooled sensitivity and specificity were 0.27 (95% CI 0.24–0.30) and 0.94 (95% CI 0.92–0.96), respectively [22].
On further analysis of each study, the detection of cHPV-DNA in plasma in early-stage cervical cancer showed mixed results, with two studies showing no detection at stage 1B [9,23], three studies with <10% detection at stage 1B [10,13,24], and another three studies showing detection between 24 and 100% [12,22,25]. The studies that described high levels of cHPV-DNA in stage 1 tumours also noted that the viral DNA load was directly proportional to the clinical stage; the patients also tended to be high-risk, i.e., lymphovascular space invasion (LVSI), deep stromal invasion, and tumour size > 20 mm. This may go some way to explaining why many of the early-stage cancers in our study were negative for cHPV-DNA.
Fiala et al. described the challenges of using ctDNA for early-stage cancer diagnosis [26]. There is usually not enough ctDNA present in such cancers to allow diagnosis, which makes it difficult to design highly sensitive assays. They estimated that a tumour of 1 cm3, or 12.5 mm in diameter, will release enough ctDNA to represent 0.01% of all circulating DNA [27,28]. They concluded, therefore, that ctDNA could be used to detect tumours of approximately 10 mm but not smaller.
The strength of this study is that it focused on early-stage cervical cancers only. The low sensitivity for the detection of cHPV-DNA in early-stage cervical cancer may be explained by not only the small size of the tumour but also the inherent difficulties in the design of a sufficiently accurate HPV assay. The high specificity, however, does give some assurance that a positive test is more likely to be a true positive.
The limitation of this study is that we were unable to recruit the desired number of patients, particularly in the early cervical cancer cohort, due to the coronavirus pandemic, which also made follow-up difficult. The number did not meet the requirements of the power calculation. However, as only one cervical cancer sample was positive for cHPV-DNA, this likely would not have made a statistical difference. Another inherent limitation of this prospective cohort study was the difficulty of follow-up post-treatment. Again, as all but one of the baseline samples were negative, follow-up of these patients is unlikely to have added a significant finding to this study. Of the patients that were followed up, there were no discrepancies between baseline and follow-up samples, i.e., none became positive after treatment having had a negative baseline result. Most importantly, the sole positive patient was followed up and found to be negative after treatment.

5. Conclusions

Based on this study, we cannot recommend the use of HPVdetect as a biomarker for the diagnosis of early-stage cervical cancers, given the low sensitivity demonstrated. However, we have shown that circulating HPV is not detectable in pre-invasive disease, supporting the hypothesis that such lesions do not shed DNA into the circulation. Patients with pre-invasive disease have an effective screening tool based on high-risk HPV detection and cytology in cervical samples. Whilst work is ongoing on the use of HPVdetect as a potential biomarker for monitoring treatment effectiveness and predicting recurrence in locally advanced cervical cancers, further research is needed to establish whether any cDNA assay has sufficient sensitivity for reliable detection of early-stage cervical cancer.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers15092590/s1, Figure S1: Bioanalyzer traces for 9 libraries; Figure S2: Amplification plots from qPCR of the libraries; Table S1: Cost calculation.

Author Contributions

S.J.B.: project administration, writing original draft, data curation, investigation, formal analysis; J.L.: investigation; R.G.: investigation, manuscript review and editing; A.J.: investigation, manuscript review and editing; A.O.: resources, supervision, manuscript review and editing; A.N.R.: resources, supervision, manuscript review and editing; R.J.C.: resources, formal analysis; I.G.-M.: resources; N.T.: resources, investigation; formal analysis; S.L.: conceptualisation, visualization, manuscript review and editing; S.A.B.: conceptualisation, supervision, project administration, manuscript review. All authors have read and agreed to the published version of the manuscript.

Funding

Part of this work was undertaken by The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research and was funded by the joint Royal Marsden and Imperial confidence in concept grant (ICiC5). The Royal Marsden NHS Foundation Trust received a proportion of its funding from the NHS Executive; the views expressed in this publication are those of the authors and not necessarily those of the NHS Executive. This work and sample processing, which was performed by the RMH Biobank, were supported by the National Institute for Health Research, Royal Marsden, the Institute of Cancer Research Biomedical Research Centre, and the Clinical Research Facility BRC grant no. A67.

Institutional Review Board Statement

This study was reviewed and approved by the Committee for Clinical Research (Review Board) at the Royal Marsden Hospital NHS Trust (Ref: CCR 4157). This study was also reviewed and approved by the Health Research Authority (HRA) North-East—York Research Ethics Committee (REC)—Ref-14/NE/1055.

Informed Consent Statement

All participants provided written informed consent for participation in this study.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors acknowledge the support of Cancer Research UK Programme Grants C46/A10588, C7224/A13407, C7224/A23275, and C7224/A28724 (RAdNet). A.N.R. was supported by the NIHR Biomedical Research Centre at University College London Hospitals, the National Health Service Foundation Trust, and University College London. The authors also acknowledge The Scottish Human Papillomavirus Investigators Network (SHINe) and The Scottish HPV Archive for providing tissue samples for validation.

Conflicts of Interest

S.J.B., R.G., J.L., A.O., A.R., A.J., I.J.C., R.C., and N.T. have no declarations. S.L. declares honaria from M.S.D., G.S.K., and Elekta. S.B. declares honoraria from Elekta.

References

  1. Cancer Research UK. Cancer Research UK. [Online]. Available online: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/cervical-cancer#heading-Zero (accessed on 11 May 2018).
  2. Brown, K.; Rumgay, H.; Dunlop, C.; Ryan, M.; Quartly, F.; Cox, A.; Deas, A.; Elliss-Brookes, L.; Gavin, A.; Hounsome, L.; et al. The fraction of cancer attributable to modifiable risk factors in England, Wales, Scotland, Northern Ireland, and the United Kingdom in 2015. Br. J. Cancer 2018, 118, 1130–1141. [Google Scholar] [CrossRef] [PubMed]
  3. Green, J.; de Gonzalez, A.B.; Sweetland, S.; Beral, V.; Chilvers, C.; Crossley, B.; Deacon, J.; Hermon, C.; Jha, P.; Mant, D.; et al. Risk factors for adenocarcinoma and squamous cell carcinoma of the cervix in women aged 20–44 years: The UK national case-control study of cervical cancer. Br. J. Cancer 2003, 89, 2078–2086. [Google Scholar] [CrossRef]
  4. Bosch, X.; de Sanjose, S. The epidemiology of human papillomavirus infection and cervical cancer. Dis. Markers 2007, 23, 213–227. [Google Scholar] [CrossRef] [PubMed]
  5. International Agency for Research on Cancer. List of Classifications by Cancer Sites with Sufficient or Limited Evidence in Humans, IARC Monographs Volumes 1—128a. November 2020. [Online]. Available online: https://monographs.iarc.fr/agents-classified-by-the-iarc/ (accessed on 2 January 2021).
  6. Rykova, E.Y.; Morozkin, E.S.; Ponomaryova, A.A.; Loseva, E.M.; Zaporozhchenko, I.A.; Cherdyntseva, N.V.; Vlassov, V.V.; Laktionov, P.P. Cell-free and cell-bound circulating nucleic acid complexes: Mechanisms of generation, concentration and content. Expert Opin. Biol. Ther. 2012, 12 (Suppl. 1), S141–S153. [Google Scholar] [CrossRef] [PubMed]
  7. Thierry, A.; El Messaoudi, S.; Gahan, P.; Stroun, M. Origins, structures, functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef]
  8. Widschwendter, A.; Blassnig, A.; Wiedemair, A.; Muller-Holzner, E.; Muller, H.; Marth, C. Human papillomavirus DNA in sera of cervical cancer patients as tumor marker. Cancer Lett. 2003, 202, 231–239. [Google Scholar] [CrossRef]
  9. Pornthanakasem, W.; Shotelersuk, K.; Termrungruanglert, W.; Voravud, N.; Niruthisard, S.; Mutirangura, A. Human papillomavirus DNA in plasma of patients with cervical cancer. BMC Cancer 2001, 1, 2. [Google Scholar] [CrossRef]
  10. Sathish, N.; Abraham, P.; Peedicayil, A.; Sridharan, G.; John, S.; Shaji, R.; Chandy, G. HPV DNA in plasma of patients with cervical carcinoma. J. Clin. Virol. 2004, 31, 204–209. [Google Scholar] [CrossRef]
  11. Damerla, R.R.; Lee, N.Y.; You, D.; Soni, R.; Shah, R.; Reyngold, M.; Katabi, N.; Wu, V.; McBride, S.M.; Tsai, C.J.; et al. Detection of early human papillomavirus-associated cancers by liquid biopsy. JCO Precis. Oncol. 2019, 3, 1–17. [Google Scholar] [CrossRef]
  12. Kang, Z.; Stevanovic, S.; Hinrichs, C.; Cao, L. Circulating cell-free DNA for metastatic cervical cancer detection, genotyping, and monitoring. Clin. Cancer Res. 2017, 23, 6856–6862. [Google Scholar] [CrossRef]
  13. Dong, S.; Pai, S.; Rha, S.; Hildesheim, A.; Kurman, R.; Schwartz, P. Detection and quantitation of human papillomavirus DNA in the plasma of patients with cervical carcinoma. Cancer Epidemiol. Biomark. Prev. 2002, 11, 3–6. [Google Scholar]
  14. Lee, J.Y.; Garcia-Murillas, I.; Cutts, R.J.; De Castro, D.G.; Grove, L.; Hurley, T.; Wang, F.; Nutting, C.; Newbold, K.; Harrington, K.; et al. Predicting response to radical (chemo) radiotherapy with circulating HPV DNA in locaally advanced head and neck squamous carcinoma. Br. J. Cancer 2017, 117, 876–883. [Google Scholar] [CrossRef]
  15. Lee, J.; Cutts, R.; White, I.; Augustin, Y.; Garcia-Murillas, I.; Fenwick, K.; Matthews, N.; Turner, N.; Harrington, K.; Gilbert, D.; et al. Next generation sequencing assay for detection of circulating HPV DNA (cHPV-DNA) in patients undergoing radical (chemo)radiotherapy in anal squamous cell carcinoma (ASCC). Front. Oncol. 2020, 10, 505. [Google Scholar] [CrossRef]
  16. Garcia-Murrillas, I.; Schiavon, G.; Weigelt, B.; Ng, C.; Hrebien, S.; Cutts, R. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci. Transl. Med. 2015, 7, 302ra133. [Google Scholar] [CrossRef]
  17. Quinlan, A.; Hall, I. BEDTools: A flexible suite of utilities for comparing genomic features. Bioinformatics 2010, 26, 841–842. [Google Scholar] [CrossRef]
  18. Li, H.; Handsaker, B.; Wysoker, A.; Fennell, T.; Ruan, J.; Homer, N.; Marth, G.; Abecasis, G.; Durbin, R. The sequence alignment/map format and SAMtools. Bioinformatics 2009, 25, 2078–2079. [Google Scholar] [CrossRef]
  19. Trigg, R.M.; Martinson, L.J.; Parpart-Li, S.; Shaw, J.A. Factors that influence quality and yield of circulating-free DNA: A systematic review of the methodology literature. Heliyon 2018, 4, e00699. [Google Scholar] [CrossRef]
  20. Lalondrelle, S.; Lee, J.; Cutts, R.J.; Garcia Murillas, I.; Matthews, N.; Turner, N.; Harrington, K.; Vroobel, K.; Moretti, E.; Bhide, S.A. Predicting Response to Radical Chemoradiotherapy with Circulating HPV DNA (cHPV-DNA) in Locally Advanced Uterine Cervix Cancer. Cancers 2023, 15, 1387. [Google Scholar] [CrossRef]
  21. Gu, Y.; Wan, C.; Qiu, J.; Cui, Y.; Jiang, T.; Zhuang, Z. Circulating CHPV-DNA in the blood as a reliable biomarker for cervical cancer: A meta-analysis. PLoS ONE 2020, 15, e0224001. [Google Scholar] [CrossRef]
  22. Jeannot, E.; Becette, V.; Campitelli, M.; Calmejane, M.; Lappartient, E.; Ruff, E.; Saada, S.; Holmes, A.; Bellet, D.; Sastre-Garau, X. Circulating human papillomavirus DNA detected using droplet digital PCR in the serum of patients diagnosed with early stage human papillomavirus-associated invasive carcinoma. J. Pathol. Clin. Res. 2016, 2, 201–209. [Google Scholar] [CrossRef]
  23. Campitelli, M.; Jeannot, E.; Peter, M.; Lappartient, E.; Saada, S.; de la Rochefordière, A.; Fourchotte, V.; Alran, S.; Petrow, P.; Cottu, P.; et al. Human papillomavirus mutational insertion: Specific marker of circulating tumor DNA in cervical cancer patients. PLoS ONE 2012, 7, e43393. [Google Scholar] [CrossRef] [PubMed]
  24. Jaberipour, M.; Samsami, A.; Sahraiian, F.; Kazerooni, T.; Hashemi, M.; Ghaderi, A. Elevation of HPV-18 and HPV-16 DNA in the plasma of patients with advanced cervical cancer. Asian Pac. J. Cancer Prev. APJCP 2011, 12, 163–167. [Google Scholar] [PubMed]
  25. Hsu, K.; Huang, S.; Hsiao, J.; Cheng, Y.; Wang, S.; Chou, C. Clinical significance of serum human papillomavirus DNA in cervical carcinoma. Obstet. Gynecol. 2003, 102, 1344–1351. [Google Scholar] [CrossRef]
  26. Fiala, C.; Kulasingam, V.; Diamandis, E.P. Circulating tumor DNA for early cancer detection. J. Appl. Lab. Med. 2018, 3, 300–313. [Google Scholar] [CrossRef]
  27. Diamandis, E.; Fiala, C. Can circulating tumor DNA be used for direct and early stage cancer detection? F1000Research 2017, 6, 2129. [Google Scholar] [CrossRef] [PubMed]
  28. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef]
Table 1. Patient demographics.
Table 1. Patient demographics.
Demographics
AgeN (%)
25–3534 (65)
36–4513 (25)
46–553 (6)
>552 (4)
Ethnicity
White British32 (62)
European9 (17)
Asian5 (10)
Black2 (4)
Other4 (7)
Smoker
Previous11 (21)
Current16 (31)
No25 (48)
HPV Subtype
168 (40)
183 (15)
Non 16/189 (45)
Table 2. Stage of cancer and grade of pre-invasive lesion.
Table 2. Stage of cancer and grade of pre-invasive lesion.
ResultsStageNumber of PatientsTotal
Cancers1A1612
1B14
2B2
Pre-invasiveCIN 1940
CIN 217
CIN 311
CGIN3
Table 3. Stage of cancer and grade of pre-invasive lesion, with HPV subtype (where known) and number of amplicons read. * Excluded from study.
Table 3. Stage of cancer and grade of pre-invasive lesion, with HPV subtype (where known) and number of amplicons read. * Excluded from study.
Sample No.DiagnosisHPV Type CytologyHPV Type PlasmaNumber of Amplicons > 6Pos/Neg
421A1-161NEG
441A1-521NEG
521A1--0NEG
241A116-0NEG
251A1181811POS
351A116161NEG
411B1--0NEG
101B1-161NEG
231B1-312NEG
22B181879POS *
42BNON 16/18-0NEG *
5CGIN-161NEG
31CGIN--0NEG
40CGIN--0NEG
49CIN 116-0NEG
1CIN 1--0NEG
8CIN 1NON 16/18163NEG
17CIN 118331NEG
21CIN 1--0NEG
22CIN 1--0NEG
28CIN 116-0NEG
37CIN 1--0NEG
38CIN 1--0NEG
43CIN 2--0NEG
45CIN 2--0NEG
46CIN 2-161NEG
47CIN 2--0NEG
51CIN 2--0NEG
3CIN 2NON 16/18-0NEG
6CIN 2NON 16/18-0NEG
7CIN 216-0NEG
12CIN 2NON 16/18-0NEG
18CIN 2--0NEG
19CIN 2-162NEG
20CIN 2NON 16/18-0NEG
29CIN 2NON 16/18-0NEG
34CIN 2-161NEG
36CIN 2-330NEG
39CIN 2--0NEG
48CIN 3--0NEG
50CIN 3--0NEG
13CIN 316161NEG
14CIN 316-0NEG
15CIN 316-0NEG
16CIN 3--0NEG
26CIN 3--0NEG
27CIN 3-160NEG
30CIN 3--0NEG
32CIN 3--0NEG
33CIN 3-160NEG
Table 4. Follow-up sample HPV data.
Table 4. Follow-up sample HPV data.
Sample No.Initial DiagnosisHPV Type PlasmaHPV Type f/uNumber of Amplicons > 6Pos/Neg
441A152-0NEG
3CIN 2Non 16/18162NEG
5CGIN16-0NEG
6CIN 2-161NEG
7CIN 2--0NEG
8CIN 11616/183NEG
91B1No sample183NEG
101B116161NEG
11CIN 2No sample-0NEG
12CIN 2--0NEG
13CIN 316-0NEG
16CIN 3-331NEG
18CIN 2-331NEG
21CIN 1--0NEG
231B131181NEG
241A1-181NEG
251A118161NEG
31CGIN--0NEG
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bryan, S.J.; Lee, J.; Gunu, R.; Jones, A.; Olaitan, A.; Rosenthal, A.N.; Cutts, R.J.; Garcia-Murillas, I.; Turner, N.; Lalondrelle, S.; et al. Circulating HPV DNA as a Biomarker for Pre-Invasive and Early Invasive Cervical Cancer: A Feasibility Study. Cancers 2023, 15, 2590. https://doi.org/10.3390/cancers15092590

AMA Style

Bryan SJ, Lee J, Gunu R, Jones A, Olaitan A, Rosenthal AN, Cutts RJ, Garcia-Murillas I, Turner N, Lalondrelle S, et al. Circulating HPV DNA as a Biomarker for Pre-Invasive and Early Invasive Cervical Cancer: A Feasibility Study. Cancers. 2023; 15(9):2590. https://doi.org/10.3390/cancers15092590

Chicago/Turabian Style

Bryan, Stacey J., Jen Lee, Richard Gunu, Allison Jones, Adeola Olaitan, Adam N. Rosenthal, Ros J. Cutts, Isaac Garcia-Murillas, Nick Turner, Susan Lalondrelle, and et al. 2023. "Circulating HPV DNA as a Biomarker for Pre-Invasive and Early Invasive Cervical Cancer: A Feasibility Study" Cancers 15, no. 9: 2590. https://doi.org/10.3390/cancers15092590

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop