Next Article in Journal
Relationship between Prognostic Nutritional Index and Amputation in Patients with Diabetic Foot Ulcer
Next Article in Special Issue
A Multichannel CT and Radiomics-Guided CNN-ViT (RadCT-CNNViT) Ensemble Network for Diagnosis of Pulmonary Sarcoidosis
Previous Article in Journal
An Integrated Machine Learning Approach for Congestive Heart Failure Prediction
Previous Article in Special Issue
Machine Learning Approach for Improved Longitudinal Prediction of Progression from Mild Cognitive Impairment to Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Radiomics Features in Predicting Human Papillomavirus Status in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review, Quality Appraisal, and Meta-Analysis

by
Golnoosh Ansari
1,
Mohammad Mirza-Aghazadeh-Attari
2,*,
Kristine M. Mosier
3,
Carole Fakhry
4 and
David M. Yousem
5
1
Department of Radiology, Northwestern Hospital, Northwestern School of Medicine, Chicago, IL 60611, USA
2
Division of Interventional Radiology, Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
3
Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
4
Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
5
Division of Neuroradiology, Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
*
Author to whom correspondence should be addressed.
Diagnostics 2024, 14(7), 737; https://doi.org/10.3390/diagnostics14070737
Submission received: 20 December 2023 / Revised: 18 March 2024 / Accepted: 20 March 2024 / Published: 29 March 2024
(This article belongs to the Special Issue Artificial Intelligence in Clinical Decision Support)

Abstract

:
We sought to determine the diagnostic accuracy of radiomics features in predicting HPV status in oropharyngeal squamous cell carcinoma (SCC) compared to routine paraclinical measures used in clinical practice. Twenty-six articles were included in the systematic review, and thirteen were used for the meta-analysis. The overall sensitivity of the included studies was 0.78, the overall specificity was 0.76, and the overall area under the ROC curve was 0.84. The diagnostic odds ratio (DOR) equaled 12 (8, 17). Subgroup analysis showed no significant difference between radiomics features extracted from CT or MR images. Overall, the studies were of low quality in regard to radiomics quality score, although most had a low risk of bias based on the QUADAS-2 tool. Radiomics features showed good overall sensitivity and specificity in determining HPV status in OPSCC, though the low quality of the included studies poses problems for generalizability.

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is the seventh most common cancer globally, accounting for more than 300,000 annual deaths [1]. Alarmingly, it is projected that there will be a 30% increase in the overall incidence of HNSCC globally, with both developed and developing nations experiencing a growth in case numbers. This trend is driven by human papillomavirus (HPV), especially in North America and Europe [2,3], although there are other risk factors involved in the development of HNSCC such as alcohol consumption, tobacco products, blood group type, and more [4,5]. Even today, it is estimated that the frequency of HPV-positive oropharyngeal (OP) SCC is 2.5 times that of HPV-negative OPSCC [6]. Furthermore, although HPV-positive and -negative HNSCC have conventionally been classified as a single clinical diagnosis and represented as a single entity in the ICD classification [7], they show different clinical profiles and have recently been viewed as two distinct clinical entities by the National Comprehensive Cancer Network [8]. HPV-positive OPSCC usually has a higher 5-year overall, progression-free, and recurrence-free survival compared to HPV-negative OPSCC. It has also been shown that responses to conventional chemo-radiotherapy and immunotherapy are significantly different between the two groups [9,10]. Importantly, there are also critical differences between the two in tumor staging: the American Joint Committee on Cancer (AJCC) staging guidelines now offer two different T and N staging systems for OPSCC depending upon HPV status, essentially classifying them as two distinct clinical entities [11,12].
Conventionally, tissue samples have been used to document the presence of HPV in cancer cells, but recently, texture analysis features extracted from medical images have been proposed as a potential means to differentiate between HPV-positive and -negative OPSCC [13]. Radiomics features are mathematically extracted as high-throughput quantitative features that represent valuable information that is not readily/visibly appreciated by radiologists [14]. These features have the potential to substitute for tissue sampling as a form of virtual biopsy, as subtle nuances in the texture of a given lesion may be indicative of differences in microanatomy, which itself could be a reflection of different genomic alterations and pathologic processes in the cancer [15,16].
Currently, there is a lack of meta-analytic evidence regarding the possible role of radiomics features in determining HPV status in HNSCC [17]. Differences in the methodology of these studies, such as feature extraction, modality of choice, and issues regarding classification and validation, limit the generalizability of the results of isolated studies.
This investigation aims to determine the value of radiomics features extracted from medical images including CT, MRI, and ultrasound in determining HPV status in OPSCC and to determine the quality of radiomics methodologies used in the published literature.

2. Materials and Methods

2.1. Literature Search

Two researchers (G.A., M.M.A.A.) independently searched the PubMed, Scopus, Embase, and Web of Science repositories using the following combinations:
((HPV) AND (Radiomics)) AND (HNSCC), ((human papilloma) AND (Radiomics)) AND (HNSCC), ((papilloma) AND (Radiomics)) AND (head and neck), ((papilloma) AND (Radiomics)) AND (opscc), ((HPV) AND (Radiomics)) AND (opscc), ((papilloma) AND (texture)) AND (opscc), ((papilloma) AND (texture)) AND (hnscc), ((HPV) AND (texture)) AND (hnscc), (radiomics) AND (HPV), (radiomics) AND (human papillomavirus), (texture) AND (human papillomavirus).
Exclusion criteria consisted of studies that were not in English, conference papers, editorials, reports from society meetings or Kaggle challenges (open dataset challenges recruiting teams to train and validate predictive models or similar schemes) [18], abstracts, and review articles.
The same researchers were also responsible for screening the articles based on title and abstract to exclude irrelevant studies. After the suitable studies were identified, information regarding their methodology and numeric findings (namely the number of true positive, true negative, false positive, and false negative cases) were extracted. Only studies reporting complete metadata were included in the meta-analysis.

2.2. Quality Assessment

The radiomics quality score (RQS) and Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) were used by the two researchers (G.A. and M.M.A.A.) to evaluate the methodological quality and risk of bias of the studies included in the systematic review. The two readers independently graded the studies and any case of disagreement was solved via discussion until consensus was achieved.

2.3. Statistical Analysis

Statistical analyses were performed with the MIDAS package Stata software, version 16.0, and the Meta-DiSc 2.0 web application. Statistical heterogeneity was assessed using the I2 value, providing an estimate of the percentage of variability among the included reports. I2 values of 0–25%, 25–50%, 50–75%, and >75% represented very low, low, medium, and high heterogeneity, respectively.
Pooled sensitivity, specificity, diagnostic odds ratio (DOR), positive and negative likelihood ratios (PLR and NLR), and area under curve (AUC), with corresponding 95% confidence intervals (CIs), were calculated. A forest plot and summary receiver operating characteristic (SROC) plot were generated for each of the modalities and in total. The pooling of studies and effect size were evaluated using a random-effects model, and sub-group analysis was performed to assess the diagnostic accuracy of radiomics features extracted from each imaging modality used.
Deek’s funnel plot was used to determine publication bias, and Fagan’s nomogram was used to determine clinical utility.

3. Results

3.1. Characteristics of the Included Studies

Twenty-six studies were included in the systematic review. Of these studies, only 14 studies containing 21 databases reported complete diagnostic results, which were included in the meta-analysis. One study was later excluded from the analysis due to reporting results obtained from patients with different types of HNPCC. A PRISMA flow chart of the study is presented in Figure 1. Table 1 Presents more information about the studies included in the study. OPSCC was by far the most common head and neck cancer studied, as 23 of the studies in the systematic review only included patients with OPSCC. Two studies included a variety of HNSCCs, and a single study included non-OPSCC HNSCCs. Only studies including OPSCC were included in the meta-analysis. Contrast-enhanced CT was the most prevalent modality used for texture feature extraction (sixteen studies), followed by MRI (eight studies using different combinations of sequences used for extraction of features), and PET-CT (two studies). The earliest study we were able to find was published in 2015, with 16 studies published between 2020 and 2023.

3.2. Methodological Quality

The QUADAS-2 tool was used to determine the methodologic qualities of the included manuscripts in the systematic review (Supplementary Table S1). Generally, most of the studies had a low risk of bias in patient selection, index testing, and timing. However, there was an undetermined risk of bias regarding the reference test, as different studies utilized different methods to determine if a lesion was HPV positive, including the detection of HPV DNA, HPV messenger RNA (mRNA), or the p16 protein. Polymerase chain reaction has inherent limitations in determining HPV infection, with the most important being the fact that PCR cannot differentiate low-risk strains of HPV from transcriptionally active ones, which are shown to be involved in the pathogenesis of OPSCC. radiomics quality score of the included articles is presented in Supplementary Table S2 and Figure 1.

3.3. Publication Bias

Figure 2 depicts the Deek’s plot of asymmetry and the funnel plot of the study. There was no significant publication bias among the publications included in the meta-analysis.

3.4. Diagnostic Accuracy of Radiomics MRI

Twenty-one datasets pertaining to 14 studies were initially sought to be included in the meta-analysis. Of these studies, 13 studies incorporating 19 datasets exclusively included OPSCC cases, while a single study [29] did not determine the exact origin of the lesions included in their investigation. There were 1104 HPV-positive cancers and 748 HPV-negative lesions included. The pre-test probability of having the condition equaled 59%.
Figure 3 and Figure 4 depict the forest plot for sensitivity and specificity of radiomics features in determining HPV status in HNSCCs. The overall sensitivity of the included studies was 0.78 (0.74, 0.82), and the overall specificity was 0.76 (0.71, 0.81). The diagnostic odds ratio (DOR) equaled 12 (8, 17).
Subgroup analysis was performed based on modality, and the relative sensitivity level for MRI vs. CT comparison was 0.9 (sensitivity of 0.7, 0.8, respectively, 0.8–1.0, p = 0.27), and the relative specificity level for MRI vs. CT comparison was 1.0 (specificity of 0.79, 0.73, respectively, 0.9–1.1, p = 0.5) showed no significant difference between the two modalities. The total accuracy of datasets including MRI was 0.76, and the total accuracy of the studies including CT was also 0.76. Figure 5 presents the SROC curve of the studies based on the two modalities. It is evident that a lesser degree of variability was seen for results pertaining to datasets that used CT imaging.
Summary findings for a pre-test HPV-positive prevalence of 59% are presented in Figure 6.

3.5. Heterogeneity Assessment

The I2 statistic showed that heterogeneities for sensitivity and specificity were medium (I2 = 56.9% and 55.4%, respectively).

3.6. Clinical Utility

Figure 7 depicts the Fagan nomogram. Using a radiomics model generated on cross-sectional imaging would increase the post-test probability to 83% from 58%, with a positive likelihood ratio of 3.0 when the pre-test was positive. When the pre-test was negative, the post-test probability decreased to 29% with a negative likelihood ratio of 0.3.

4. Discussion

In the present review, we present meta-analytic evidence regarding the possible role of radiomics features extracted from CT and MR in determining the status of HPV in oropharyngeal squamous cell carcinoma. Our results show that the combined sensitivity and specificity of texture features equaled 78% and 76%, respectively, with studies using CT imaging showing less variability in their results. Interestingly, the following descriptive characteristics and their quantitative representative texture features were consistently observed to differentiate between HPV-positive and -negative lesions: HPV-positive lesions were more homogenous, spherical, and smaller, contained fewer clusters of areas with low HU (probably due to less tissue necrosis) [45], and showed lower ADC values.
Methodologically, most studies included patients from a single center for the development of their model, did not perform external validation, and acquired low radiomics quality scores. Furthermore, there are methodologic nuances that further limit the extent to which the results could be generalizable to different clinical settings: one crucial issue being the lack of utilization of Image Biomarker Standardization Initiative (IBSI)-approved extraction tools and IBSI-approved nomenclature for texture features of interest [46,47]. This is of particular importance since the next step in the implementation of radiomics to this population is the employment of established radiomics classifiers on external datasets and validation of competing classifiers, both of which could highly benefit from established IBSI performance benchmarks [29,48,49]. For example, some of the studies included in our meta-analysis utilized cases from an already established dataset on the Cancer Imaging Archive, which could serve as an opportunity for collaborative model development and external benchmarking of trained models [50,51]. Another potential factor that could limit the generalizability of the results included in the meta-analysis and introduce a means of heterogeneity is the differences in inclusion criteria of patients, especially the inclusion of patients in different stages of OPSCC. HPV-positive stage III-IV OPSCC lesions may have more histologic and anatomic similarity to HPV-negative lesions compared to lower-stage HPV-positive OPSCCs. This particular variation in patient inclusion may result in overly optimistic results in studies including a larger cohort of patients with earlier-stage OPSCC. This is of particular importance as most of the included studies in the meta-analysis did not report data regarding the stages of the included tumors, increasing their potential risk of bias in patient selection. Furthermore, of the 14 studies included in the meta-analysis, 13 exclusively focused on OPSCC, while a single study did not determine the exact origin of the lesions included, though the majority of their patient cohort consisted of patients with OPSCC (more than 75%) [41].
HPV is a well-established biomarker of prognosis. Routine HPV testing for oropharynx cancers is indeed recommended in NCCN guidelines, and clinical trials also incorporate HPV status with de-intensification trials focusing on HPV-positive patients [8]. Furthermore, the NCCN and updated American Joint Commission on Cancer guidelines consider them separate clinical conditions. This growing clinical importance of determining the status of HPV in HNSCC has led multi-disciplinary researchers to develop and validate surrogate markers that can readily determine high-risk infection [52]. One of the most promising developments has been the introduction of “NavDX® liquid biopsy for the detection of circulating tumor-modified HPV DNA [53]. The lack of consensus regarding the definition of a clear cut-off for a positive immunohistochemical (IHC) test and concerns regarding varying sensitivity and specificity rates of this method arising from technical considerations have further necessitated a revisit of diagnostic approaches [54]. Other possible supplements to IHC have been the utilization of polymerase chain reaction (PCR) and in situ hybridization to detect HPV DNA [52,55,56]. However, these new methods may face barriers to feasibility and wide-scale application [13].
Since medical imaging of HNSCC and OPSCC is an essential part of staging the tumor and determining subsequent medical or surgical treatment plans, radiomics texture features offer a potential solution to characterizing the HPV status of these tumors. Our results show that the pooled sensitivity and specificity of radiomics features (77.2% and 76.3%) were below that of the currently established methods mentioned above, as IHC has shown a sensitivity and specificity close to 90% [57]. Though the current pieces of evidence do not suggest a substitutive role for radiomics features instead of routine para-clinical practices such as IHC, they provide insight regarding the additive value of radiomics features to these established diagnostic methods. Furthermore, artificial intelligence algorithms seem to be improving with each successive month, and, as more cases of HPV-positive and -negative cancers are accumulated, it is likely that radiomics features will become more reliable and accurate. This may be true not only for HPV status but also for prognostication and predicting response to treatment without regard to the HPV status of the lesion [58]. Future investigations should leverage the existing publicly available datasets and classifiers to develop, train, and externally validate new features [59]. These endeavors should also take into account clinical variables such as tumor grading and staging in developing predictive models and investigate the possible associations between characteristics such as blood types (recognized as risk factors for hypopharyngeal and oral cavity cancers) and clinical course of HNSCCs [60,61]. Future studies should also focus on increasing the interpretability of models by introducing feature maps and SHAP summary plots [62].

5. Conclusions

The present meta-analysis showed a combined sensitivity and specificity of 77.2% and 76%, respectively, for radiomics features in determining HPV status in OPSCC. The accuracy of radiomics features extracted from imaging is lower than that of the already established para-clinical IHC methods. However, since CT and MR imaging are essential in the work-up of most OPSCCs, collaborative learning models may uncover untapped potential for radiomics features in the determination of HPV status, prediction of treatment response, and other important factors in the diagnosis and therapy of OPSCC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/diagnostics14070737/s1, Table S1. Quality of the included studies based on QUADAS-2 tool. Table S2. Radiomics Quality Score of the included studies.

Author Contributions

Conceptualization, G.A., D.M.Y. and M.M.-A.-A.; methodology, M.M.-A.-A. and G.A.; software, G.A.; validation, D.M.Y., K.M.M. and C.F.; formal analysis, M.M.-A.-A. and G.A.; investigation, G.A.; resources, D.M.Y.; data curation, G.A.; writing—original draft preparation, G.A. and D.M.Y.; writing—review and editing, C.F. and K.M.M.; visualization, M.M.-A.-A.; supervision, D.M.Y.; project administration, D.M.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The IRB of the institute waived the need for an IRB as no HIPPA-protected data were used and the study did not include individual patient data.

Informed Consent Statement

No patients were included in the study.

Data Availability Statement

All data will be made available upon request to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  2. Liao, C.-I.; Francoeur, A.A.; Kapp, D.S.; Caesar, M.A.P.; Huh, W.K.; Chan, J.K. Trends in Human Papillomavirus–Associated Cancers, Demographic Characteristics, and Vaccinations in the US, 2001–2017. JAMA Netw. Open 2022, 5, e222530. [Google Scholar] [CrossRef] [PubMed]
  3. Chaturvedi, A.K.; Engels, E.A.; Pfeiffer, R.M.; Hernandez, B.Y.; Xiao, W.; Kim, E.; Jiang, B.; Goodman, M.T.; Sibug-Saber, M.; Cozen, W.; et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J. Clin. Oncol. 2011, 29, 4294–4301. [Google Scholar] [CrossRef] [PubMed]
  4. Singh, K.; Kote, S.; Patthi, B.; Singla, A.; Singh, S.; Kundu, H.; Jain, S. Relative Risk of Various Head and Neck Cancers Among Different Blood Groups: An Analytical Study. J. Clin. Diagn. Res. 2014, 8, ZC25–ZC28. [Google Scholar] [CrossRef] [PubMed]
  5. Jaleel, B.F.; Nagarajappa, R. Relationship between ABO blood groups and oral cancer. Indian J. Dent. Res. 2012, 23, 7–10. [Google Scholar] [CrossRef]
  6. Tran, K.B.; Lang, J.J.; Compton, K.; Xu, R.; Acheson, A.R.; Henrikson, H.J.; Kocarnik, J.M.; Penberthy, L.; Aali, A.; Abbas, Q.; et al. The global burden of cancer attributable to risk factors, 2010–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2022, 400, 563–591. [Google Scholar] [CrossRef] [PubMed]
  7. Vigneswaran, N.; Williams, M.D. Epidemiologic Trends in Head and Neck Cancer and Aids in Diagnosis. Oral Maxillofac. Surg. Clin. N. Am. 2014, 26, 123–141. [Google Scholar] [CrossRef]
  8. Caudell, J.J.; Gillison, M.L.; Maghami, E.; Spencer, S.; Pfister, D.G.; Adkins, D.; Birkeland, A.C.; Brizel, D.M.; Busse, P.M.; Cmelak, A.J.; et al. NCCN Guidelines® Insights: Head and Neck Cancers, Version 1.2022. J. Natl. Compr. Cancer Netw. 2022, 20, 224–234. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, J.; Sun, H.; Zeng, Q.; Guo, X.-J.; Wang, H.; Liu, H.-H.; Dong, Z.-Y. HPV-positive status associated with inflamed immune microenvironment and improved response to anti-PD-1 therapy in head and neck squamous cell carcinoma. Sci. Rep. 2019, 9, 13404. [Google Scholar] [CrossRef]
  10. Kobayashi, K.; Hisamatsu, K.; Suzui, N.; Hara, A.; Tomita, H.; Miyazaki, T. A Review of HPV-Related Head and Neck Cancer. J. Clin. Med. 2018, 7, 241. [Google Scholar] [CrossRef]
  11. Amin, M.B.; Edge, S.B. AJCC Cancer Staging System; American College of Surgeons: Washington, DC, USA, 2020. [Google Scholar]
  12. Zanoni, D.K.; Patel, S.G.; Shah, J.P. Changes in the 8th Edition of the American Joint Committee on Cancer (AJCC) Staging of Head and Neck Cancer: Rationale and Implications. Curr. Oncol. Rep. 2019, 21, 52. [Google Scholar] [CrossRef]
  13. Augustin, J.G.; Lepine, C.; Morini, A.; Brunet, A.; Veyer, D.; Brochard, C.; Mirghani, H.; Péré, H.; Badoual, C. HPV Detection in Head and Neck Squamous Cell Carcinomas: What Is the Issue? Front. Oncol. 2020, 10, 1751. [Google Scholar] [CrossRef] [PubMed]
  14. Ardakani, A.A.; Bureau, N.J.; Ciaccio, E.J.; Acharya, U.R. Interpretation of radiomics features–A pictorial review. Comput. Methods Programs Biomed. 2022, 215, 106609. [Google Scholar] [CrossRef]
  15. Caruso, D.; Polici, M.; Zerunian, M.; Del Gaudio, A.; Parri, E.; Giallorenzi, M.A.; De Santis, D.; Tarantino, G.; Tarallo, M.; di Accadia, F.M.D.; et al. Radiomic Cancer Hallmarks to Identify High-Risk Patients in Non-Metastatic Colon Cancer. Cancers 2022, 14, 3438. [Google Scholar] [CrossRef]
  16. Shui, L.; Ren, H.; Yang, X.; Li, J.; Chen, Z.; Yi, C.; Zhu, H.; Shui, P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front. Oncol. 2020, 10, 570465. [Google Scholar] [CrossRef] [PubMed]
  17. Spadarella, G.; Ugga, L.; Calareso, G.; Villa, R.; D’aniello, S.; Cuocolo, R. The impact of radiomics for human papillomavirus status prediction in oropharyngeal cancer: Systematic review and radiomics quality score assessment. Neuroradiology 2022, 64, 1639–1647. [Google Scholar] [CrossRef]
  18. Tolkachev, A.; Sirazitdinov, I.; Kholiavchenko, M.; Mustafaev, T.; Ibragimov, B. Deep Learning for Diagnosis and Segmentation of Pneumothorax: The Results on the Kaggle Competition and Validation Against Radiologists. IEEE J. Biomed. Health Inform. 2021, 25, 1660–1672. [Google Scholar] [CrossRef]
  19. Boot, P.A.; Mes, S.W.; de Bloeme, C.M.; Martens, R.M.; Leemans, C.R.; Boellaard, R.; van de Wiel, M.A.; de Graaf, P. Magnetic resonance imaging based radiomics prediction of Human Papillomavirus infection status and overall survival in oropharyngeal squamous cell carcinoma. Oral Oncol. 2023, 137, 106307. [Google Scholar] [CrossRef]
  20. Bos, P.; Brekel, M.W.v.D.; Taghavi, M.; Gouw, Z.A.; Al-Mamgani, A.; Waktola, S.; Aerts, H.J.; Beets-Tan, R.G.; Castelijns, J.A.; Jasperse, B. Largest diameter delineations can substitute 3D tumor volume delineations for radiomics prediction of human papillomavirus status on MRI’s of oropharyngeal cancer. Phys. Medica. 2022, 101, 36–43. [Google Scholar] [CrossRef] [PubMed]
  21. Bagher-Ebadian, H.; Siddiqui, F.; I Ghanem, A.; Zhu, S.; Lu, M.; Movsas, B.; Chetty, I.J. Radiomics outperforms clinical factors in characterizing human papilloma virus (HPV) for patients with oropharyngeal squamous cell carcinomas. Biomed. Phys. Eng. Express 2022, 8, 045010. [Google Scholar] [CrossRef]
  22. Park, Y.M.; Lim, J.; Koh, Y.W.; Kim, S.; Choi, E.C. Machine learning and magnetic resonance imaging radiomics for predicting human papilloma virus status and prognostic factors in oropharyngeal squamous cell carcinoma. Head Neck 2022, 44, 897–903. [Google Scholar] [CrossRef] [PubMed]
  23. Lv, W.; Xu, H.; Han, X.; Zhang, H.; Ma, J.; Rahmim, A.; Lu, L. Context-Aware Saliency Guided Radiomics: Application to Prediction of Outcome and HPV-Status from Multi-Center PET/CT Images of Head and Neck Cancer. Cancers 2022, 14, 1674. [Google Scholar] [CrossRef] [PubMed]
  24. Marzi, S.; Piludu, F.; Avanzolini, I.; Muneroni, V.; Sanguineti, G.; Farneti, A.; D’urso, P.; Benevolo, M.; Rollo, F.; Covello, R.; et al. Multifactorial Model Based on DWI-Radiomics to Determine HPV Status in Oropharyngeal Squamous Cell Carcinoma. Appl. Sci. 2022, 12, 7244. [Google Scholar] [CrossRef]
  25. Sohn, B.; Choi, Y.S.; Ahn, S.S.; Kim, H.; Han, K.; Lee, S.; Kim, J. Machine learning based radiomic HPV phenotyping of oropharyngeal SCC: A feasibility study using MRI. Laryngoscope 2021, 131, E851–E856. [Google Scholar] [CrossRef] [PubMed]
  26. Bagher-Ebadian, H.; Siddiqui, F.; Ghanem, A.; Zhu, S.; Lu, M.; Movsas, B.; Chetty, I. Superiority of Radiomics Information Compared to Clinical Factors in Characterization of Human Papilloma Virus (HPV) Status in Patients with Oropharyngeal Squamous Cell Carcinomas. Int. J. Radiat. Oncol. Biol. Phys. 2021, 111, e405–e406. [Google Scholar] [CrossRef]
  27. Song, B.; Yang, K.; Garneau, J.; Lu, C.; Li, L.; Lee, J.; Stock, S.; Braman, N.M.; Koyuncu, C.F.; Toro, P.; et al. Radiomic Features Associated with HPV Status on Pretreatment Computed Tomography in Oropharyngeal Squamous Cell Carcinoma Inform Clinical Prognosis. Front. Oncol. 2021, 11, 744250. [Google Scholar] [CrossRef] [PubMed]
  28. Bagher-Ebadian, H.; Lu, M.; Siddiqui, F.; Ghanem, A.I.; Wen, N.; Wu, Q.; Liu, C.; Movsas, B.; Chetty, I.J. Application of radiomics for the prediction of HPV status for patients with head and neck cancers. Med. Phys. 2020, 47, 563–575. [Google Scholar] [CrossRef] [PubMed]
  29. Bogowicz, M.; Jochems, A.; Deist, T.M.; Tanadini-Lang, S.; Huang, S.H.; Chan, B.; Waldron, J.N.; Bratman, S.; O’sullivan, B.; Riesterer, O.; et al. Privacy-preserving distributed learning of radiomics to predict overall survival and HPV status in head and neck cancer. Sci. Rep. 2020, 10, 4542. [Google Scholar] [CrossRef] [PubMed]
  30. Bos, P.; Brekel, M.W.M.v.D.; Gouw, Z.A.R.; Al-Mamgani, A.; Waktola, S.; Aerts, H.J.W.L.; Beets-Tan, R.G.H.; Castelijns, J.A.; Jasperse, B. Clinical variables and magnetic resonance imaging-based radiomics predict human papillomavirus status of oropharyngeal cancer. Head Neck 2020, 43, 485–495. [Google Scholar] [CrossRef]
  31. Choi, Y.; Nam, Y.; Jang, J.; Shin, N.-Y.; Ahn, K.-J.; Kim, B.-S.; Lee, Y.-S.; Kim, M.-S. Prediction of Human Papillomavirus Status and Overall Survival in Patients with Untreated Oropharyngeal Squamous Cell Carcinoma: Development and Validation of CT-Based Radiomics. Am. J. Neuroradiol. 2020, 41, 1897–1904. [Google Scholar] [CrossRef]
  32. Haider, S.P.; Mahajan, A.; Zeevi, T.; Baumeister, P.; Reichel, C.; Sharaf, K.; Forghani, R.; Kucukkaya, A.S.; Kann, B.H.; Judson, B.L.; et al. PET/CT radiomics signature of human papilloma virus association in oropharyngeal squamous cell carcinoma. Eur. J. Nucl. Med. 2020, 47, 2978–2991. [Google Scholar] [CrossRef] [PubMed]
  33. Ren, J.; Yuan, Y.; Qi, M.; Tao, X. Machine learning–based CT texture analysis to predict HPV status in oropharyngeal squamous cell carcinoma: Comparison of 2D and 3D segmentation. Eur. Radiol. 2020, 30, 6858–6866. [Google Scholar] [CrossRef] [PubMed]
  34. Suh, C.H.; Lee, K.H.; Choi, Y.J.; Chung, S.R.; Baek, J.H.; Lee, J.H.; Yun, J.; Ham, S.; Kim, N. Oropharyngeal squamous cell carcinoma: Radiomic machine-learning classifiers from multiparametric MR images for determination of HPV infection status. Sci. Rep. 2020, 10, 17525. [Google Scholar] [CrossRef] [PubMed]
  35. Mungai, F.; Verrone, G.B.; Pietragalla, M.; Berti, V.; Addeo, G.; Desideri, I.; Bonasera, L.; Miele, V. CT assessment of tumor heterogeneity and the potential for the prediction of human papillomavirus status in oropharyngeal squamous cell carcinoma. La Radiol. Medica 2019, 124, 804–811. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, C.; Cintra, M.; Brennan, K.; Zhou, M.; Colevas, A.D.; Fischbein, N.; Zhu, S.; Gevaert, O. Development and validation of radiomic signatures of head and neck squamous cell carcinoma molecular features and subtypes. EBioMedicine 2019, 45, 70–80. [Google Scholar] [CrossRef] [PubMed]
  37. Lee, J.Y.; Han, M.; Kim, K.S.; Shin, S.-J.; Choi, J.W.; Ha, E.J. Discrimination of HPV status using CT texture analysis: Tumour heterogeneity in oropharyngeal squamous cell carcinomas. Neuroradiology 2019, 61, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  38. Leijenaar, R.T.; Bogowicz, M.; Jochems, A.; Hoebers, F.J.; Wesseling, F.W.; Huang, S.H.; Chan, B.; Waldron, J.N.; O’Sullivan, B.; Rietveld, D.; et al. Development and validation of a radiomic signature to predict HPV (p16) status from standard CT imaging: A multicenter study. Br. J. Radiol. 2018, 91, 20170498. [Google Scholar] [CrossRef] [PubMed]
  39. Ranjbar, S.; Ning, S.; Zwart, C.M.; Wood, C.P.; Weindling, S.M.; Wu, T.; Mitchell, J.R.; Li, J.; Hoxworth, J.M. Computed tomography-based texture analysis to determine human papillomavirus status of oropharyngeal squamous cell carcinoma. J. Comput. Assist. Tomogr. 2018, 42, 299–305. [Google Scholar] [CrossRef]
  40. Ravanelli, M.; Grammatica, A.; Tononcelli, E.; Morello, R.; Leali, M.; Battocchio, S.; Agazzi, G.; Bastia, M.B.d.M.e.; Maroldi, R.; Nicolai, P.; et al. Correlation between Human Papillomavirus Status and Quantitative MR Imaging Parameters including Diffusion-Weighted Imaging and Texture Features in Oropharyngeal Carcinoma. Am. J. Neuroradiol. 2018, 39, 1878–1883. [Google Scholar] [CrossRef]
  41. Bogowicz, M.; Riesterer, O.; Ikenberg, K.; Stieb, S.; Moch, H.; Studer, G.; Guckenberger, M.; Tanadini-Lang, S. Computed Tomography Radiomics Predicts HPV Status and Local Tumor Control After Definitive Radiochemotherapy in Head and Neck Squamous Cell Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 921–928. [Google Scholar] [CrossRef]
  42. Yu, K.; Zhang, Y.; Yu, Y.; Huang, C.; Liu, R.; Li, T.; Yang, L.; Morris, J.S.; Baladandayuthapani, V.; Zhu, H. Radiomic analysis in prediction of Human Papilloma Virus status. Clin. Transl. Radiat. Oncol. 2017, 7, 49–54. [Google Scholar] [CrossRef] [PubMed]
  43. Fujita, A.; Buch, K.; Li, B.; Kawashima, Y.; Qureshi, M.M.; Sakai, O. Difference between HPV-positive and HPV-negative non-oropharyngeal head and neck cancer: Texture analysis features on CT. J. Comput. Assist. Tomogr. 2016, 40, 43–47. [Google Scholar] [CrossRef] [PubMed]
  44. Buch, K.; Fujita, A.; Li, B.; Kawashima, Y.; Qureshi, M.M.; Sakai, O. Using texture analysis to determine human papillomavirus status of oropharyngeal squamous cell carcinomas on CT. Am. J. Neuroradiol. 2015, 36, 1343–1348. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, Y.H.; Kim, Y.S.; Chung, M.J.; Yu, M.; Jung, S.L.; Yoo, I.R.; Lee, Y.S.; Kim, M.S.; Sun, D.I.; Kang, J.H. Soft Tissue Necrosis in Head and Neck Cancer Patients After Transoral Robotic Surgery or Wide Excision With Primary Closure Followed by Radiation Therapy. Medicine 2016, 95, e2852. [Google Scholar] [CrossRef] [PubMed]
  46. Zwanenburg, A.; Vallières, M.; Abdalah, M.A.; Aerts, H.J.W.L.; Andrearczyk, V.; Apte, A.; Ashrafinia, S.; Bakas, S.; Beukinga, R.J.; Boellaard, R.; et al. The image biomarker standardization initiative: Standardized quantitative radiomics for high-throughput image-based phenotyping. Radiology 2020, 295, 328–338. [Google Scholar] [CrossRef] [PubMed]
  47. Hatt, M.; Vallieres, M.; Visvikis, D.; Zwanenburg, A. IBSI: An international community radiomics standardization initiative. J. Nucl. Med. 2018, 59 (Suppl. 1), 287. [Google Scholar]
  48. Rieke, N.; Hancox, J.; Li, W.; Milletarì, F.; Roth, H.R.; Albarqouni, S.; Bakas, S.; Galtier, M.N.; Landman, B.A.; Maier-Hein, K.; et al. The future of digital health with federated learning. NPJ Digit. Med. 2020, 3, 119. [Google Scholar] [CrossRef]
  49. Pati, S.; Baid, U.; Edwards, B.; Sheller, M.; Wang, S.-H.; Reina, G.A.; Foley, P.; Gruzdev, A.; Karkada, D.; Davatzikos, C.; et al. Federated learning enables big data for rare cancer boundary detection. Nat. Commun. 2022, 13, 7346. [Google Scholar] [CrossRef] [PubMed]
  50. Kwan, J.Y.Y.; Su, J.; Huang, S.H.; Ghoraie, L.S.; Xu, W.; Chan, B.; Yip, K.W.; Giuliani, M.; Bayley, A.; Kim, J.; et al. Radiomic biomarkers to refine risk models for distant metastasis in HPV-related oropharyngeal carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1107–1116. [Google Scholar] [CrossRef]
  51. Leijenaar, R.T.H.; Carvalho, S.; Hoebers, F.J.P.; Aerts, H.J.W.L.; van Elmpt, W.J.C.; Huang, S.H.; Chan, B.; Waldron, J.N.; O’sullivan, B.; Lambin, P. External validation of a prognostic CT-based radiomic signature in oropharyngeal squamous cell carcinoma. Acta Oncol. 2015, 54, 1423–1429. [Google Scholar] [CrossRef]
  52. Kim, K.Y.; Lewis, J.S., Jr.; Chen, Z. Current status of clinical testing for human papillomavirus in oropharyngeal squamous cell carcinoma. J. Pathol. Clin. Res. 2018, 4, 213–226. [Google Scholar] [CrossRef] [PubMed]
  53. Gunning, A.; Kumar, S.; Williams, C.K.; Berger, B.M.; Naber, S.P.; Gupta, P.B.; Fitz, C.D.V.; Kuperwasser, C. Analytical Validation of NavDx, a cfDNA-Based Fragmentomic Profiling Assay for HPV-Driven Cancers. Diagnostics 2023, 13, 725. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, Z.W.; Weinreb, I.; Kamel-Reid, S.; Perez-Ordoñez, B. Equivocal p16 immunostaining in squamous cell carcinoma of the head and neck: Staining patterns are suggestive of HPV status. Head Neck Pathol. 2012, 6, 422–429. [Google Scholar] [CrossRef] [PubMed]
  55. Fauzi, F.H.; Hamzan, N.I.; Ab Rahman, N.; Suraiya, S.; Mohamad, S. Detection of human papillomavirus in oropharyngeal squamous cell carcinoma. J. Zhejiang Univ. B 2020, 21, 961–976. [Google Scholar] [CrossRef] [PubMed]
  56. Gargano, S.M.; Sebastiano, C.; Mardekian, J.; Solomides, C.C.; HooKim, K. Cytological interpretation of p16 immunohistochemistry in head and neck carcinomas: Does the choice of fixative matter? J. Am. Soc. Cytopathol. 2021, 10, 216–224. [Google Scholar] [CrossRef] [PubMed]
  57. Hibbert, J.; Halec, G.; Baaken, D.; Waterboer, T.; Brenner, N. Sensitivity and Specificity of Human Papillomavirus (HPV) 16 Early Antigen Serology for HPV-Driven Oropharyngeal Cancer: A Systematic Literature Review and Meta-Analysis. Cancers 2021, 13, 3010. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, M.H.; Cao, D.; Ginat, D.T. Radiomic Model Predicts Lymph Node Response to Induction Chemotherapy in Locally Advanced Head and Neck Cancer. Diagnostics 2021, 11, 588. [Google Scholar] [CrossRef] [PubMed]
  59. Grossberg, A.J.; Mohamed, A.S.R.; Elhalawani, H.; Bennett, W.C.; Smith, K.E.; Nolan, T.S.; Williams, B.; Chamchod, S.; Heukelom, J.; Kantor, M.E.; et al. Imaging and clinical data archive for head and neck squamous cell carcinoma patients treated with radiotherapy. Sci. Data 2018, 5, 1. [Google Scholar] [CrossRef]
  60. Alexandra, G.; Alexandru, M.; Stefan, C.F.; Petruta-Maria, D.; Gabriel, B.M.; Dragos-Eugen, G.; Teodor, G.M. Blood Group Type Association with Head and Neck Cancer. Hematol. Rep. 2022, 14, 24–30. [Google Scholar] [CrossRef]
  61. Kakava, K.; Karelas, I.; Koutrafouris, I.; Damianidis, S.; Stampouloglou, P.; Papadakis, G.; Xenos, A.; Krania, F.; Sarof, P.; Tasopoulos, G.; et al. Relationship between ABO blood groups and head and neck cancer among Greek patients. J. BUON Off. J. Balk. Union Oncol. 2016, 21, 594–596. [Google Scholar]
  62. Ye, J.-Y.; Fang, P.; Peng, Z.-P.; Huang, X.-T.; Xie, J.-Z.; Yin, X.-Y. A radiomics-based interpretable model to predict the pathological grade of pancreatic neuroendocrine tumors. Eur. Radiol. 2023, 34, 1994–2005. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA diagram of the study, showing the included studies.
Figure 1. PRISMA diagram of the study, showing the included studies.
Diagnostics 14 00737 g001
Figure 2. (A) Funnel plot with pseudo 95% confidence interval and (B) Deek’s funnel plot of asymmetry of the included studies. The two plots show that although there was some heterogeneity witnessed on the funnel plot, there was no significant publication bias in the included articles.
Figure 2. (A) Funnel plot with pseudo 95% confidence interval and (B) Deek’s funnel plot of asymmetry of the included studies. The two plots show that although there was some heterogeneity witnessed on the funnel plot, there was no significant publication bias in the included articles.
Diagnostics 14 00737 g002
Figure 3. Forest plot showing the individual sensitivity of the included studies and their respective datasets in the meta-analysis [20,21,22,23,24,31,32,33,34,35,36,37,38].
Figure 3. Forest plot showing the individual sensitivity of the included studies and their respective datasets in the meta-analysis [20,21,22,23,24,31,32,33,34,35,36,37,38].
Diagnostics 14 00737 g003
Figure 4. Forest plot showing the individual specificity of the included studies and their respective datasets in the meta-analysis [20,21,22,23,24,31,32,33,34,35,36,37,38].
Figure 4. Forest plot showing the individual specificity of the included studies and their respective datasets in the meta-analysis [20,21,22,23,24,31,32,33,34,35,36,37,38].
Diagnostics 14 00737 g004
Figure 5. SROC curve of the studies included in the meta-analysis based on their utilized modality. The red line and the circles show studies using CT, and the green lines and triangles show studies using MRI. CT imaging showed a lesser degree of variation in diagnostic accuracy results compared to MR imaging.
Figure 5. SROC curve of the studies included in the meta-analysis based on their utilized modality. The red line and the circles show studies using CT, and the green lines and triangles show studies using MRI. CT imaging showed a lesser degree of variation in diagnostic accuracy results compared to MR imaging.
Diagnostics 14 00737 g005
Figure 6. Summary findings of the meta-analysis, based on a pre-test prevalence of 58% and a hypothetical sample size of 100.
Figure 6. Summary findings of the meta-analysis, based on a pre-test prevalence of 58% and a hypothetical sample size of 100.
Diagnostics 14 00737 g006
Figure 7. Fagan nomogram depicting the clinical utility of radiomics features in determining HPV status in the datasets included in the meta-analysis.
Figure 7. Fagan nomogram depicting the clinical utility of radiomics features in determining HPV status in the datasets included in the meta-analysis.
Diagnostics 14 00737 g007
Table 1. Characteristics of the articles included in the systematic review. Bold articles are those that are also included in the meta-analysis. Bold feature extraction software are those adherent to IBSI nomenclature.
Table 1. Characteristics of the articles included in the systematic review. Bold articles are those that are also included in the meta-analysis. Bold feature extraction software are those adherent to IBSI nomenclature.
CountryYearStudy Name Retro/ProCancer ModalityNumber of Features Used for Model Development/Considered Significant Extracted Features, Number Incorporated into Model Name of Features Included in the Model (If Available/Applicable) Technique Used for Feature Selection Classification Method Segmentation Feature Extraction Seg SoftwareMajor Texture/Biologic Correlate (If Any)
The Netherlands 2022Boot et al. [19]Retrospective Oropharyngeal squamous cell carcinomaPre-treatment T1 20498Run entropy, 10th percentile, dependence count energy, compactness, center of mass shift, minor axis length, asphericity, Zone distance non-uniformity GLDZM, flatness, high dependence, low gray level emphasis, coefficient of variation, and quartile coefficient RFE–random forest, RFE–logistic regressionRandom forest, logistic regressionManual/semiautomatic RaCat softwVelocity AI 3.1HPV-negative tumors are more heterogenous, while HPV-positive lesions are more spherical and have a clear defined margin
The Netherlands2022Bos et al. [20]Retrospective OPSCCT1W MRI20-9 1184Not determined individuallyICC + Mann–Whitney ULogistic regression predictionManual PyRadiomics3D slicer software2D texture features could be used in lieu of 3D volume-of-interest features in differentiating HPV-negative and -positive lesions.
United States2022Bagher-Ebadian [21]Retrospective OPSCCContrast-enhanced
computed tomography
14192Morphology, IBS,
GLSZM, GLDZM, NGLDM, GLCM, GLRLM
Non-parametric tests, correlation analysis, LASSO Logistic regression Manual ROdiomiXROdiomiXTumors of HPV+ patients are smaller and more spherical compared to the HPV- patients. Compared to HPV- patients, the absolute magnitude of HU of tumors in HPV+ patients is higher
Republic of Korea 2022Park et al. [22]Retrospective OPSCCT1 Gd-enhanced axial images8140Skewness, maximum, 10 percentile, minimum surface volume ratio, coarseness, correlation, lmc2, contrast1 LASSOLogistic regression model and a light gradient boosting machine (LightGBM)Manual Pyradiomics3D slicerIn HPV-positive OPSCC, tumor boundaries were more clear and showed cystic changes in lymph node metastasis. It was also reported that the minimum apparent diffusion coefficient of HPV-positive OPSCC was remarkably low
China 2022Wenbing et al. [23]Retrospective OPSCC PET/CT 7112Un-named radiomics features selected based on saliency maps AUC of each feature Multivariate logistic regression modelSemi-manual SERA; Ver 2.1SERA; Ver 2.1High saliency values were more frequently located at the remote region relative to the centroid for both PET and CT images, indicating the aggressiveness and special biological behavior of the tumor periphery
Italy2022Marzi [24]Retrospective OPSCCMRI diffusion map 215750th percentile of DPT, inverse difference
Moment of ADCLN
Minimum redundancy maximum relevance (MRMR)Naive Bayes classificationManual IBEX3D slicer HPV-positive tumors had a marked reduction in ADC values and a more leptokurtic and skewed right ADC histogram
Republic of Korea2021Sohn [25]Retrospective OPSCCT1WI and fat-saturated6170firstorder_Skewness, glcm_Imc1, ngtdm_Coarseness, firstorder_Skewness, ngtdm_Strength, shape_FlatnessLASSO Based on coefficients of LASSO Manual Pyradiomics 2.1.03D slicerN/A
United States 2021Bagher-Ebadian [26]Retrospective OPSCCCE-CT15192Major axis length, GLSZ—zone size non-uniformity, root mean square intensity, minor axis length, GLDZM—small distance low gray level emphasis, NGLDM—high dependence high gray level emphasis, GLCM—information correlation, GLCM—correlation, MORPH—flatness, GLRLM—long run low gray level emphasis, GLDZM—large zone low gray level emphasis, GLSZM—large zone low gray level emphasis, NGLDM—dependence count variance, IH—maximum histogram intensityLASSOFiltered LASSO-GLM classifierManual ROdiomiXROdiomiXIntensity-based features (IVH, IH, IBS) became significantly different between HPV-negative and -positive groups in the post-frequency filtered datasets, suggesting a possible role of frequency analysis
United States2021Song et al. [27]Retrospective OPSCCCE-CT15454Kurtosis of diff-average, skewness of sum-average, median, λ = 3, θ = 0.1 rad, skewness, λ = 5, θ = 0.0 rad MRMRLASSO Score Manual In-house-developed N/AHigher
Haralick correlation shows less pixel intensity
disorders and decreased morphologic appearance heterogeneity
for HPV-positive tumors compared with HPV-negative ones
USA2020Ebadian et al. [28]Retrospective OPSCCPre-treatment CE-CT14172DOST- ENERGY-1, DOST- ENERGY-2, DOST- ENERGY-3, DOST- ENERGY-6, DOST- ENERGY-7, DOST- ENERGY-8, DOST- ENERGY-9, GLCM—contrast, GLCM—energy, GLCM—entropy, mean breadth 13, spherical disproportionLevene KS test absolute biserial correlation GLM Manual MatLabMatLab HPV+ tumors have a lower component density compared to the HPV– patients.
HPV+ tumors have higher image contrast, with images being patchier than HPV-negative tumors.
HPV+ tumors are more simple, symmetric, and more rounded
Switzerland2020Bogowicz et al. [29]Retrospective HNSCC (6 cohorts)Pre-treatment CE-CT10 centralized/12 distributed 981HLH GLCM cluster shade, LHH GLCM cluster shade, HLH 10th percentile, HHL GLCM cluster shade, LLH NGLDM Idhge, LHH GLSZM sze, HLL GLDZM idlge, asphericirty, LLH GLSZM SZE, HLL GLSZM SZE, LHL GLSZM sze ICC + the average linkage hierarchical clustering + univariate logistic regressionMultivariate logistic regression modelManual Z-Rad radiomics softwareZ-Rad radiomics softwareN/A
The Netherlands2020Bos et al. [30]Retrospective OPSCCPostcontrast 3DT1W MRIIn radiomics model 3, 14 combined, 11 unique features overall DistributedShape sphericity, gray level co-occurrence matrix inverse
difference moment (Laplacian of Gaussian), kurtosis (wavelet)
Recursive feature elimination, ICC, Man WiU Logistic regressionManual PyRadiomics 2.2.0Not determined Shape sphericity, gray level co-occurrence matrix inverse difference moment (Laplacian of Gaussian, kurtosis)
Republic of Korea 2020Y Choi [31]Retrospective OPSCC Contrast-enhanced CT9854original_shape_Flatness
original_shape_SphericalDisproportion
wavelet_HLH_firstorder_Mean
wavelet_HLH_firstorder_Uniformity
wavelet_HLH_glcm_ClusterShade
wavelet_LHH_glcm_Idm
wavelet_LHH_glcm_Imc1
wavelet_LHL_glszm_SmallAreaHighGrayLevelEmphasis wavelet_LLH_glcm_Imc2
Boruta (random forest-based wrapper algorithm)None Manual PyRadiomicssyngo.via frontier software (Siemens)Spatial heterogeneity of tumors might be associated with patients’ prognosis
United States 2020Stefan P Haider [32]Retrospective OPSCC PET/CT 30, 21, 9 2074N/AVariousXGB, SVM, RF, NB, EINETManual, separately on PET and CT Pyradiomcis3D slicerRadiomics signatures based on primary tumor lesions had higher predictive performance compared to those based on metastatic cervical lymph nodes; however, models using consensus VOI of all nodes had better classification performance than individual nodes
China2020Ren [33]Retrospective OPSCCCT6,786Inverse variance, gray level nonuniformity, inverse variance, energy, dependence variance, informational measure of correlation 1, informational measure of correlation 2, inverse variance, correlation, median, inverse variance, correlation, gray level nonuniformityICC, wrapper-based algorithm k-nearest neighbors (k-NN), logistic regression (LR), and random forest (RF)Manual PyRadiomics 2.2.03D slicerN/A
Republic of Korea2020Suh [34]Retrospective OPSCCT1WI, T2WI, CE-T1WI, and ADC maps2211618Not determined individually LASSOLogistic regression, random forestManual MatlabMedical
imaging interaction toolkit (MITK)
Features extracted from ADC maps were the most relevant in differentiating HPV-positive and -negative lesions
Italy2019Mungai [35]Retrospective OPSCCContrast-enhanced CT imaging1342LRE, SRHGE, LRLGE, LRHGE, GLNU, coarseness, contrast, busyness, LZE, LZLGE, LZHGE, GLNUIndependent samples t-test N/AManual (LIFEx)Syngo.via, SiemensN/A
China2019Huang et al. [36]Retrospective OPSCC CE-CT5540 firstOrder_InterquartileRange, waveletHHH_firstOrder_Kurtosis, waveletLLL_firstOrder_Maximum, shapeSize_Solidity, firstOrder_QuartileCoefficientDispersionMinimum redundancy maximum relevance (mRMR), LASSO Logistic regression Manual Matlab 2018aMatlab 2018aN/A
Republic of Korea 2019Ji Young Lee et al. [37]Retrospective OPSCCContrast-enhanced CTN/A6Mean, SD, entropy, MPP, skewness, kurtosisN/ANone Manual TexRAD,TexRAD,N/A
The Netherlands2018Leijenar [38]Retrospective OPSCCContrast-enhanced CT imaging165,173902Laplacian of Gaussian (3 mm) kurtosis, gray level size zone matrix,
low gray level large size emphasis, Laplacian of Gaussian (4 mm)
10th percentile, gray level co-occurrence matrix inverse variance, gray level size zone matrix, small zone emphasis
LASSOMultivariable logistic regressionManual Matlab 2014aN/AHigher homogeneity in HPV-positive group—lower-contrast uptake
United States2018Ranjbar [39]Retrospective OPSCC Contrast-enhanced neck CT377Median and entropy and GLCM entropyIndependent samples t-test Diagonal quadratic
discriminant analysis
Manual OsiriXOsiriXDifference in texture features could reflect the different amounts of water and keratinization in tumors
Italy 2018Ravanelli [40]Retrospective OPSCCT1, T2, DWI 830Mean ADC, MPP T2 SSF 4, MPP T2 SSF 3, mean T2 SSF 2, mean T2 SSF 3, mean T2 SSF 1, mean T2 SSF 4, kurtosis VIBE SSF 4 Mann–Whitney U test or Student t-testN/AManual TexRADTexRADHPV-positive tumors had a significantly lower ADC than HPV-negative tumors
Switzerland2017Bogowicz et al. [41]Retrospective Stage III and IV HNSCCPre-treatment CE-CT43171. LLL standard deviation
2. LLL small-zone high gray level emphasis
3. HHL difference entropy
4. Coefficient of variation
Principal component analysis + Horn methodMultivariable Cox regression analysisManual In-house-developed PythonIn-house-developed PythonMore homogeneity, fewer small regions with high intensity
United States 2017Yu et al. [42]Retrospective OPSCCCT21683Mean breadth and
spherical disproportion
Wilcoxon rank-sum test, AUC of each future alone Logistic regressionManual IBEXIBEXHPV-positive patients usually have smaller and simpler tumors
United States 2016Fujita et al. [43]Retrospective None OPSCC HNC Contrast-enhanced CT16425 histogram, 3 gray level co-occurrence matrix), 1 gray level run-length feature, 2 gray level gradient matrix features, and 5 law features None Manual MATLABAW workstationSpatially dependent variations in features could be used to represent histologic differences in HPV-positive and -negative tumors
United States 2015Buch et al. [44]Retrospective OPSCC Contrast-enhanced CT342Entropy, median, GLCM texture feature entropyStudent t-test None Manual In-house-developed Matlab AW workstation (GE Healthcare)N/A
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

Ansari, G.; Mirza-Aghazadeh-Attari, M.; Mosier, K.M.; Fakhry, C.; Yousem, D.M. Radiomics Features in Predicting Human Papillomavirus Status in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review, Quality Appraisal, and Meta-Analysis. Diagnostics 2024, 14, 737. https://doi.org/10.3390/diagnostics14070737

AMA Style

Ansari G, Mirza-Aghazadeh-Attari M, Mosier KM, Fakhry C, Yousem DM. Radiomics Features in Predicting Human Papillomavirus Status in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review, Quality Appraisal, and Meta-Analysis. Diagnostics. 2024; 14(7):737. https://doi.org/10.3390/diagnostics14070737

Chicago/Turabian Style

Ansari, Golnoosh, Mohammad Mirza-Aghazadeh-Attari, Kristine M. Mosier, Carole Fakhry, and David M. Yousem. 2024. "Radiomics Features in Predicting Human Papillomavirus Status in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review, Quality Appraisal, and Meta-Analysis" Diagnostics 14, no. 7: 737. https://doi.org/10.3390/diagnostics14070737

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