Next Article in Journal
Ten Issues for Updating in Community-Acquired Pneumonia: An Expert Review
Previous Article in Journal
Larval Wound Therapy: Possibilities and Potential Limitations—A Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

p16 Overexpression in Sinonasal Squamous Cell Carcinoma: Association with Human Papillomavirus and Prediction of Survival Outcomes

1
Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara-cho, Nakagami-gun, Okinawa 903-0215, Japan
2
Department of Pathology and Oncology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara-cho, Nakagami-gun, Okinawa 903-0215, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2023, 12(21), 6861; https://doi.org/10.3390/jcm12216861
Submission received: 28 September 2023 / Revised: 26 October 2023 / Accepted: 27 October 2023 / Published: 30 October 2023
(This article belongs to the Section Otolaryngology)

Abstract

:
p16 overexpression is often used as a surrogate marker for human papillomavirus (HPV) infection in oropharyngeal squamous cell carcinoma but remains an uncertain diagnostic tool for HPV-related sinonasal squamous cell carcinoma (SNSCC). Our study involved 79 consecutive SNSCC patients who were treated at a tertiary referral university hospital during 2006–2021. We retrospectively examined their clinical characteristics and conducted p16 immunohistochemistry and HPV detection. We found that 12.7% of the patients exhibited p16 overexpression, which was significantly more common in the nasal cavity and increased from 2015 onward. The HPV was a high-risk type and viral loads ranged from 4.2 to 1.6 × 106 copies/ng DNA with genome integration. Five-year overall survival (OS) and five-year relapse-free survival (RFS) rates were 74.6% and 69.9%, respectively. Our multivariate analysis showed that T category (T1–4a) and hemoglobin levels (≥13.7) were significant favorable prognostic factors for OS, while T category, performance status, and p16 overexpression were significantly associated with RFS. In patients with p16 overexpression, OS was 100% and RFS was 90%. Our findings suggest that p16 overexpression is a reliable surrogate marker for transcriptionally active HPV infection and predicts a favorable prognosis.

Graphical Abstract

1. Introduction

Sinonasal malignancies, although infrequent, account for about 3–5% of all head and neck cancers [1]. From 2004 to 2008, the annual incidence rate for sinonasal cancer was between 5 and 10 cases per million in males and 2 and 5 cases per million in females [2]. Sinonasal squamous cell carcinoma (SNSCC) is the predominant histological subtype, comprising 35–60% of all cases [3]. Notably, the incidence of sinonasal cancer exhibits marked regional variations, with the incidence of sinonasal malignancies appearing to be relatively high in the Asia–Pacific region compared with Europe and North America [2]. Radical surgery is the first choice of treatment, but alternative treatment approaches have been sought because of challenges such as aesthetic considerations, ethnic diversity, and the difficulty of securing clear surgical margins.
High-risk human papillomavirus (HPV) types are well-established culprits in oropharyngeal squamous cell carcinoma (OPSCC) [4]. In recent years, these high-risk HPV strains have been posited as potential drivers of SNSCC onset [3,5,6,7,8]. According to recent meta-analyses, the HPV prevalence in SNSCC cases is around 25.5% [5]. Moreover, HPV status seems to have a tangible correlation with survival outcomes, exhibiting modest heterogeneity across studies [8]. p16-positive malignant tumors in the larynx as well as OPSCC demonstrated an improved survival outcome after treatment [9]. Nevertheless, there is a wide range of reported HPV infection rates in SNSCC, owing to methodological differences and the limited samples in each study. Notably, the geographic variability in HPV prevalence seen in SNSCC mirrors the patterns seen in OPSCC [6], adding complexity in pinning down a direct association between high-risk HPV strains and SNSCC. In the diagnostic realm, p16 immunohistochemistry is favored for its cost-effectiveness and broad applicability, making it the go-to method for identifying transcriptionally active HPV infection in OPSCC [10]. However, p16 immunoreactivity is not a consistent marker for transcriptionally active high-risk HPV infection in sinonasal samples [11].
This study investigated in detail the diagnostic precision of p16 immunohistochemistry for detecting HPV DNA. Additionally, we aimed to shed light on the clinical features and survival outcomes associated with HPV-related SNSCC.

2. Materials and Methods

2.1. Study Design

In this retrospective observational study, we reviewed the clinical records of patients diagnosed with SNSCC (both keratinizing and nonkeratinizing SCC according to the update in the 5th edition of the WHO classification [12]) between August 2006 and November 2021 at a single tertiary referral university hospital (Ryukyu University Hospital). From this pool, 79 consecutive patients with SNSCC who underwent surgery or chemoradiotherapy (CRT) with curative intent were selected. We meticulously excluded patients with non-SCC tumor histology. Furthermore, we also excluded patients receiving palliative care due to distant metastasis or general complications and those who opted for the best supportive care (Figure 1). SNSCC consisted of 59% of all sinonasal malignant tumors.
All participants provided written informed consent before the collection and analysis of tumor samples. This study was approved by the Institutional Review Board of the University of the Ryukyus (project ID: 156) and complied with the ethical guidelines of the 1975 Declaration of Helsinki, as revised in 2008.

2.2. Clinical Evaluation

A multidisciplinary team, consisting of head and neck surgeons, radiation oncologists, and reconstructive surgeons, participated in pre-treatment evaluation and decision-making. This evaluation entailed an exhaustive review of the patient’s medical history, physical examination, performance status assessment, serum chemistry profiles, complete blood cell count, chest X-ray, computed tomography (CT), and magnetic resonance imaging (MRI). Positron emission tomography combined with CT was performed to identify lymph nodes or distant metastases.
Data on clinical parameters and oncological outcomes were recorded up to the end of the follow-up period on 31 July 2023. We determined clinical and pathological tumor staging in line with the Union for International Cancer Control (UICC) TNM Classification (eighth edition, 2017) [13].
Several parameters for nutritional status and general condition were assessed before treatment, including hemoglobin (Hb), albumin (Alb), neutrophil–lymphocyte ratio (NLR) [14], C-reactive protein (CRP), geriatric nutritional risk index (GNRI) [15], modified Glasgow prognostic score (mGPS) [16], prognostic nutritional index (PNI) [17] and Eastern Cooperative Oncology Group Performance Status (ECOG PS) [18,19]. PNI is derived from serum Alb levels and lymphocyte count [20], while NLR is the ratio of absolute neutrophil count to absolute lymphocyte count. These universally employed indices gauge inflammatory and nutritional status in cancer patients.
After primary treatment, we regularly checked for locoregional recurrence and distant metastasis through physical examinations, CT and/or MRI, and head and neck endoscopic examinations.

2.3. Surgical Treatment

Patients requiring total maxillectomy underwent combined neck dissection with reconstructive surgery in addition to removal of the primary lesion. Endoscopic sinus surgery was chosen for limited primary lesions where assured surgical margins were anticipated. Typically, postoperative radiation therapy (60 Gy), supplemented with platinum infusion (100 mg/m2, two or three times triweekly), was applied to the primary site and/or neck within 6 weeks postoperatively for cases with close/positive surgical margins or extracapsular lymph node extension. In patients with renal issues and those aged over 75 years, radiation therapy was prioritized as the postoperative adjuvant treatment instead of CRT.

2.4. CRT Protocol

This protocol, which has been described previously [21], is summarized here. The irradiation focus encompassed the maxilla, ethmoid sinus, nasal cavity, and pterygopalatine fossa, delivering 70 Gy across 35 sessions over 7 weeks. Along with the primary lesion, metastatic neck lesions were also treated. At the start of intensity-modulated radiotherapy, intra-arterial chemotherapy was introduced via a femoral route. Both docetaxel (60 mg/m2) and nedaplatin (80 mg/m2) were administered to the target lesions through the deployed microcatheters two or three times every 4 weeks.

2.5. p16 Immunohistochemistry

We assessed p16 expression using the CINtec® p16 histology kit (Roche Applied Science, Penzberg, Germany) [11]. The threshold for p16 overexpression (both cytoplasmic and nuclear expression) was set as diffuse (≥75%) tumor expression with a minimum of moderate (+2/3) staining intensity, consistent with the 8th edition of the AJCC classification for OPSCC [13,22]. p16 immunoreactivity was classified according to the percentage of p16-positive cells out of all tumor cells and the intensity of p16 immunoreactivity (none, weak, moderate, or strong) (Figure 2).

2.6. HPV DNA Detection

Cases showing p16 overexpression underwent further testing by polymerase chain reaction (PCR) analysis and HPV DNA in situ hybridization. For PCR, DNA extracted from fresh frozen tumor samples was processed using the MY09/MY11 and GP5+/GP6+ degenerate consensus primer sets targeting the L1 region, as documented in previous studies [22]. The full experimental conditions are available in Supplementary Materials (Table S1).
We also introduced a novel quantitative real-time PCR assay system targeting the E6 and E2 genes of HPV strains 16, 18, 33, and 52. The assay system was designed to quantify the viral load and discern the viral integration status [11] (Table S2). For in situ hybridization analysis of high-risk HPV DNA, we employed the GenPoint HPV Biotinylated DNA Probe (Dako; Agilent Technologies, Inc., Santa Clara, CA, USA). This probe can identify an array of HPV types in formalin-fixed paraffin-embedded sections to confirm the presence of HPV DNA in SNSCC [11]. Details of the experimental conditions are presented in the Supplementary Materials.
In this research, HPV-related SNSCC was defined as cases exhibiting both p16 overexpression and the presence of HPV DNA as determined by PCR or in situ hybridization [11].

2.7. Statistical Analysis

Categorical data were analyzed using Pearson’s chi-squared test, and continuous data were analyzed using the Mann–Whitney U test. For comparisons with small sample sizes, Fisher’s exact test was used. Survival curves were plotted by the Kaplan–Meier method, and survival distributions were compared by the log-rank test. We used a multivariate Cox proportional hazard model to analyze prognostic factors and treatments influencing overall survival (OS) and relapse-free survival (RFS). Events were defined as death due to any cause for OS and as recurrence at the primary site, neck lymph node recurrence, or distant metastasis for RFS.
A p-value of <0.05 was considered statistically significant, and all analyses were performed using JMP Pro 15.0.0 (SAS Institute Inc., Cary, NC, USA).

3. Results

3.1. Patient Demographics, Clinical Testing and Treatment Profiles

The clinical characteristics of the patients are shown in Table 1. In our study cohort of seventy-nine participants, there were seventy men and nine women, with a median age of sixty-one (range: 30–90) years. Tumor locations were distributed as follows: maxillary sinus (sixty-three patients, 79.7%), ethmoid sinus (ten patients, 12.7%), and nasal cavity (six patients, 7.6%). The histologic classification was keratinizing squamous cell carcinoma in 75 patients (93.6%). There were no significant differences in tobacco and alcohol consumption, performance status, and histological differentiation between HPV-related and HPV-unrelated tumors. A notable finding was that p16 overexpression was significantly more prevalent in the nasal cavity (50.0%) than in the maxillary or ethmoid sinus (9.6%, p = 0.004, chi-squared test). A temporal trend was observed, with p16 overexpression becoming more common in patients treated after 2015 (p = 0.037, Fisher’s exact test).
Most patients were diagnosed with T4 (58.2%) and stage IV disease (68.4%). Treatment modalities included radiation-based treatment (RT/CRT) in 61 patients (77.2%), with the remaining 18 patients (22.8%) undergoing radical surgery followed by adjuvant RT/CRT-based treatment according to the pathological findings. Laboratory and nutritional variables did not show significant differences between the two tumor types (Table 2). There was no significant difference in inflammatory and nutritional status between HPV-related and HPV-unrelated tumors (Table 2).

3.2. p16 Overexpression and HPV DNA Detection

Of the 79 patients with SNSCC, 10 (12.7%) exhibited p16 overexpression. Figure 2 shows representative examples of p16 immunoreactivity. Table 3 shows the distribution of p16 immunoreactivity according to positive cell percentage and intensity. Some immunoreactivity to p16 was observed in 45.5% of the SNSCC cases, but these p16-positive cases usually had weak to moderate intensity and less than 50% positive cells (Table 3).
Every p16-overexpressing sample tested positive for high-risk HPV DNA. The distribution of HPV types included HPV-16, HPV-18, HPV-33, and HPV-52, with some unidentified types. Viral loads across samples ranged from 4.2 to 1.63 × 106 copies/ng genomic DNA, with each case displaying viral integration into the host genome (Table 4). Figure 3 shows representative images of p16 overexpression and HPV DNA in situ hybridization.

3.3. Prognostic Evaluation of Clinical Characteristics

Over a median follow-up duration of 60 months, the 5-year OS rate for the entire cohort was 74.6%. Univariate analysis identified T4b lesions and Hb levels below 13.7 g/dL as significant factors for poorer OS (p = 0.011 and p = 0.012, respectively). Although the 5-year OS rate for the p16-overexpression group was 100% (compared with 71.2% for the p16-negative group), this difference failed to achieve statistical significance by the log-rank test (p = 0.072, Figure 4).
In multivariate analysis considering the T category, Hb levels, and CRP, the significant prognostic factors for OS were T1–4a (p = 0.019) and Hb levels ≥ 13.7 g/dL (p = 0.035). Since all patients with p16 overexpression were alive, the multivariate analysis did not provide a hazard rate.
The 5-year RFS rate was 69.9%. In univariate analysis, the T category, ECOG PS, Hb levels, Alb levels, and mGPS displayed significant prognostic implications, as shown in Figure 5 and Table 5.
As visualized in Figure 5, the Kaplan-Meier curves revealed a 5-year RFS of 90% for the p16-overexpression group, compared with 66.9% for the p16-negative group. However, p16 overexpression was not significant in the univariate analysis (p = 0.142). Since these nutritional indicators confound each other, mGPS and ECOG PS were included in the multivariate analysis. In the multivariate analysis of RFS, which included the T category, Hb levels, mGPS, ECOG PS, and p16 overexpression, T1–4a (p = 0.004), ECOG PS 0 (p = 0.026), and p16 overexpression (p = 0.012) were identified as significant prognostic markers.

4. Discussion

The reported prevalence of HPV in SNSCC varies widely, ranging from 8.9% to 62% [5,23,24,25,26,27,28]. This considerable discrepancy can be attributed to differing definitions of HPV-related SNSCC, methodological differences in HPV detection, and small sample sizes. When studies focus solely on E6/E7 mRNA expression of the HPV gene, which is considered to most accurately reflect active transcriptional infection, prevalence rates tend to decrease by approximately 25% [7,25,29]. While the RNA-based test is recognized as the gold standard for accuracy, its complexity and cost prohibit its widespread use in the clinical setting [7]. Our study underscores the strong association of p16 overexpression with the presence of high-risk HPV DNA, suggesting that p16 immunohistochemistry might be a cost-effective, sensitive method for pinpointing transcriptionally active HPV infections in SNSCC. In the present study, p16 overexpression was present in 12.7% of SNSCC cases. Considering the small number of p16overexpressing cases and the cost-effectiveness of detecting HPV-related SNSCC, p16 immunohistochemistry should be examined first, and if p16 overexpression is found, PCR for HPV DNA or HPV DNA in situ hybridization tests are recommended. However, the number of patients with p16 overexpression was low (n = 10), and thus there might be a risk for bias in the present results. Further study is needed to confirm the importance of p16 overexpression in SNSCC, as shown in the present study, including the survival benefit, the characteristics of tumor location, and the increasing trend.
In OPSCC, the role of p16 overexpression as a surrogate marker for detecting active HPV infection has been well-established due to its robust predictive and prognostic value [4]. However, its precise cut-off value for overexpression in SNSCC has not yet been defined, which may account for the variance in p16 positivity rates among different studies. This lack of clarity might lead to reduced sensitivity of p16 immunohistochemistry as an indicator of transcriptionally active HPV in SNSCC as opposed to OPSCC. In the present study, approximately half of the cases had p16 immunoexpression, and moderate intensity of p16 expression was also observed in HPV-unrelated cases (Table 3). However, when the threshold for p16 overexpression was set at diffuse (≥75%) tumor expression with a minimum of moderate (+2/3) staining intensity, all samples with p16 overexpression had high-risk HPV strains with viral integration into the genome by PCR or DNA in situ hybridization, and these cases had a favorable prognosis. These results aligned with RNA-based tests, affirming their potential for effective survival prognosis stratification. In the present study, T4b and nutritional status were significantly associated with survival outcomes, which is consistent with previous reports [30,31]. Previous reports have found that low Hb and high CRP levels were associated with poor prognosis in head and neck cancer [32,33]. The results of the present study are consistent with those reports. A low Hb level has been linked to malnutrition, weight loss, and cancer cachexia [34], and a high CRP level reflects systemic inflammation. These nutritional and inflammatory markers might be important in the prognosis of SNSCC. In addition to these factors, p16 overexpression was an independent prognostic factor for RFS in multivariate analysis and linked to superior OS, as all p16-positive SNSCC of our cohort are alive. The literature has shown a possible survival advantage linked to p16 expression [24,27,28,30]. However, several reports have shown no survival benefits of p16 expression [23,35]. These discrepant findings could be attributed to differences in patient demographics, HPV testing methodologies, or classifications across studies. Our findings highlight the survival advantages of p16 overexpression in SNSCC, echoing the benefits seen in HPV-related OPSCC cases [4,22]. In the future, the definition of p16 overexpression in SNSCC should be clarified in the same way as in OPSCC through studies with larger cohorts and a unified detection protocol.
Insights into the viral load and integration status of SNSCC remain scarce. In this investigation, we provided data on HPV DNA viral load and viral integration status for SNSCC with p16 overexpression (Table 4). Our observed HPV types align with the high-risk subtypes detected in OPSCC at our institution [22]. Drawing parallels from our prior study on oropharyngeal carcinoma, the HPV-16 viral loads in OPSCC varied between 0.3 × 105 and 2.69 × 105 copies/ng DNA (median 2.666 × 104 copies/ng DNA). In contrast, the present study’s HPV-16 and HPV-33 viral loads in SNSCC ranged from 4.2 to 1.63 × 106 copies/ng DNA. Notwithstanding the limited sample size, viral loads in SNSCC did not significantly deviate from those in OPSCC, and all types showing p16 overexpression exhibited viral integration. These insights affirm the critical role of HPV, particularly in its relation to p16 overexpression, in shaping the landscape of SNSCC.
The location of HPV-related sinonasal cancer remains unclear. Previous reports showed no significant difference in HPV prevalence among sinonasal regions [7]. Recently, the prevalence of HPV was found to be higher in the nasal cavity than in the maxillary sinus by analysis of the National Cancer Database with various HPV detection methods [28]. According to a recent report from Italy [36], only 3% of nasal septum tumor was classified as SCC. In the present study, high-risk HPV was most frequently detected in the nasal cavity, which is in line with a recent meta-analysis, and this is the first report of this finding in Asia. Given the geographical and racial discrepancies in sinonasal carcinoma and HPV infection, further study is needed to clarify these aspects.
The availability of HPV status in OPSCC increased with each subsequent year of diagnosis from 2008: 0.1–0.2% for 2004–2008, 1.4% for 2009, 30.6% for 2010, 45.6% for 2011, and 56.9% for 2012 [37]. Recently, an increase in the incidence of HPV-associated SNSCC and the prevalence of HPV-positive SNSCC from 1995 to 2019 has been demonstrated using the SEER (Surveillance, Epidemiology, and End Results Program) database in the United States [38]. The results of the present study on the prevalence of HPV-related SNSCC in an Asian country are consistent with those in the United States. This might reflect the widespread HPV infection in the head and neck. We must pay attention to HPV infection in the entire head and neck region, not only the oropharynx.

5. Conclusions

p16 overexpression combined with HPV DNA testing might be a reliable surrogate marker for HPV-related SNSCC and an accurate prognostic marker. High-risk HPV infection was the most frequently detected in the nasal cavity, and the prevalence of HPV-related SNSCC is increasing in Japan as in the United States. A larger multicenter prospective study is needed to clarify the prognostic significance of high-risk HPV infection in SNSCC patients treated with a multimodal approach.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/jcm12216861/s1, Supplementary Methods; Table S1: Primers used for the detection of HPV DNA by PCR; Table S2: Primers used for the detection of viral loads and viral integration in real-time PCR.

Author Contributions

Conceptualization, H.H., T.I. and M.S.; methodology, T.I. and H.H.; software, H.H. and M.S.; validation, H.H., T.I., M.T. and M.S.; formal analysis, H.H. and M.S.; investigation, T.I., K.T., H.H. and H.K.; resources, H.H., H.K., S.K., S.A., H.M., N.K., Y.O., T.H. (Tomoyo Higa), T.H. (Teruyuki Higa), T.T. and N.W.; data curation, H.H., K.T., M.T. and M.S.; writing—original draft preparation, H.H.; writing—review and editing, M.S. and T.I.; visualization, H.H., T.I. and M.S.; project administration, M.S.; funding acquisition, H.H., T.I., M.S. and T.H. (Teruyuki Higa). All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Grants-in-Aid for Scientific Research (KAKENHI Grant Numbers 20K097581, 21K09635, 21K09585, and 23K15889, to H.H., T.I., M.S., and T.H. (Teruyuki Higa), respectively).

Institutional Review Board Statement

This study was conducted with the approval of the Institutional Review Board of the University of Ryukyus (project identification code: 156, approved in September 2007) in accordance with the 1975 Declaration of Helsinki, as revised in 2008.

Informed Consent Statement

Written informed consent was obtained from all study participants.

Data Availability Statement

The datasets generated and/or analyzed during the present study have not been made publicly available. However, data can be made available from the corresponding author upon reasonable request.

Acknowledgments

We thank Yukari Matsuda, Nanami Nakamoto, and Mami Gakiya, the Ryukyu Society for the Promotion of Oto-Rhino-Laryngology and for administrative assistance related to this investigation. In addition, we thank Narumi Hasegawa and Junko Kawakami, the Ryukyu Society for the Promotion of Oto-Rhino-Laryngology, for their support with the experiments.

Conflicts of Interest

The authors declare no conflict of interest. The sponsor had no role in the study’s design; in the collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Unsal, A.A.; Kilic, S.; Dubal, P.M.; Baredes, S.; Eloy, J.A.; EUROCARE-5 Working Group. A population-based comparison of European and North American sinonasal cancer survival. Auris Nasus Larynx 2018, 45, 815–824. [Google Scholar] [CrossRef] [PubMed]
  2. Youlden, D.R.; Cramb, S.M.; Peters, S.; Porceddu, S.V.; Møller, H.; Fritschi, L.; Baade, P.D. International comparisons of the incidence and mortality of sinonasal cancer. Cancer Epidemiol. 2013, 37, 770–779. [Google Scholar] [CrossRef] [PubMed]
  3. Tendron, A.; Classe, M.; Casiraghi, O.; Pere, H.; Even, C.; Gorphe, P.; Moya-Plana, A. Prognostic analysis of HPV status in sinonasal squamous cell carcinoma. Cancers 2022, 14, 1874. [Google Scholar] [CrossRef]
  4. Ang, K.K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tân, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef] [PubMed]
  5. Syrjänen, K.; Syrjänen, S. Detection of human papillomavirus in sinonasal carcinoma: Systematic review and meta-analysis. Hum. Pathol. 2013, 44, 983–991. [Google Scholar] [CrossRef]
  6. Chang Sing Pang, K.J.W.; Mur, T.; Collins, L.; Rao, S.R.; Faden, D.L. Human papillomavirus in sinonasal squamous cell carcinoma: A systematic review and meta-Analysis. Cancers 2020, 13, 45. [Google Scholar] [CrossRef]
  7. Svajdler, M.; Nemcova, J.; Dubinsky, P.; Metelkova, A.; Svajdler, P.; Straka, L.; Sakar, R.; Daum, O.; Michal, M.; Skalova, A.; et al. Significance of transcriptionally-active high-risk human papillomavirus in sinonasal squamous cell carcinoma: Case series and a meta-analysis. Neoplasma 2020, 67, 1456–1463. [Google Scholar] [CrossRef]
  8. Sharma, A.; Tang, A.L.; Takiar, V.; Wise-Drape, T.M. Human papillomavirus and survival of sinonasal squamous cell carcinoma patients: A systematic review and meta-analysis. Cancers 2021, 13, 3677. [Google Scholar] [CrossRef]
  9. Sireci, F.; Lorusso, F.; Dispenza, F.; Immordino, A.; Gallina, S.; Salvago, P.; Martines, F.; Bonaventura, G.; Uzzo, M.L.; Spatola, G.F. A Prospective Observational Study on the Role of Immunohistochemical Expression of Orphanin in Laryngeal Squamous Cell Carcinoma Recurrence. J. Pers. Med. 2023, 13, 1211. [Google Scholar] [CrossRef]
  10. Mehanna, H.; Taberna, M.; von Buchwald, C.; Tous, S.; Brooks, J.; Mena, M.; Morey, F.; Grønhøj, C.; Rasmussen, J.H.; Garset-Zamani, M.; et al. Prognostic implications of p16 and HPV discordance in oropharyngeal cancer (HNCIG-EPIC-OPC): A multicentre, multinational, individual patient data analysis. Lancet Oncol. 2023, 24, 239–251. [Google Scholar] [CrossRef]
  11. Hirakawa, H.; Ikegami, T.; Kise, N.; Kinjyo, H.; Kondo, S.; Agena, S.; Hasegawa, N.; Kawakami, J.; Maeda, H.; Suzuki, M. Human papillomavirus infection and EGFR exon 20 insertions in sinonasal inverted papilloma and squamous cell carcinoma. J. Pers. Med. 2023, 13, 657. [Google Scholar] [CrossRef]
  12. Agarwal, A.; Bhatt, A.A.; Bathla, G.; Kanekar, S.; Soni, N.; Murray, J.; Vijay, K.; Vibhute, P.; Rhyner, P.H. Update from the 5th Edition of the WHO Classification of Nasal, Paranasal, and Skull Base Tumors: Imaging Overview with Histopathologic and Genetic Correlation. Am. J. Neuroradiol. 2023, 44, 1116–1125. [Google Scholar] [CrossRef] [PubMed]
  13. Doescher, J.; Veit, J.A.; Hoffmann, T.K. The 8th edition of the AJCC Cancer Staging Manual: Updates in otorhinolaryngology, head and neck surgery. HNO 2017, 65, 956–961. [Google Scholar] [CrossRef] [PubMed]
  14. Ueda, T.; Chikuie, N.; Takumida, M.; Furuie, H.; Kono, T.; Taruya, T.; Hamamoto, T.; Hattori, M.; Ishino, T.; Takeno, S. Baseline neutrophil-to-lymphocyte ratio (NLR) is associated with clinical outcome in recurrent or metastatic head and neck cancer patients treated with nivolumab. Acta Otolaryngol. 2020, 140, 181–187. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, H.M.; Xu, L.; Tang, P.; Guo, R. Geriatric nutritional risk index and survival of patients with colorectal cancer: A meta-analysis. Front. Oncol. 2022, 12, 906711. [Google Scholar] [CrossRef] [PubMed]
  16. Tong, T.; Guan, Y.; Xiong, H.; Wang, L.; Pang, J. A meta-analysis of Glasgow prognostic score and modified Glasgow prognostic score as biomarkers for predicting survival outcome in renal cell carcinoma. Front. Oncol. 2020, 10, 1541. [Google Scholar] [CrossRef] [PubMed]
  17. Luan, C.W.; Tsai, Y.T.; Yang, H.Y.; Chen, K.Y.; Chen, P.H.; Chou, H.H. Pretreatment prognostic nutritional index as a prognostic marker in head and neck cancer: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 17117. [Google Scholar] [CrossRef]
  18. Chang, P.H.; Yeh, K.Y.; Huang, J.S.; Lai, C.H.; Wu, T.H.; Lan, Y.J.; Tsai, J.C.S.; Chen, E.Y.C.; Yang, S.W.; Wang, C.H. Pretreatment performance status and nutrition are associated with early mortality of locally advanced head and neck cancer patients undergoing concurrent chemoradiation. Eur. Arch. Otorhinolaryngol. 2013, 270, 1909–1915. [Google Scholar] [CrossRef]
  19. Farhangfar, A.; Makarewicz, M.; Ghosh, S.; Jha, N.; Scrimger, R.; Gramlich, L.; Baracos, V. Nutrition impact symptoms in a population cohort of head and neck cancer patients: Multivariate regression analysis of symptoms on oral intake, weight loss and survival. Oral. Oncol. 2014, 50, 877–883. [Google Scholar] [CrossRef]
  20. Bullock, A.F.; Greenley, S.L.; McKenzie, G.A.G.; Paton, L.W.; Johnson, M.J. Relationship between markers of malnutrition and clinical outcomes in older adults with cancer: Systematic review, narrative synthesis and meta-analysis. Eur. J. Clin. Nutr. 2020, 74, 1519–1535. [Google Scholar] [CrossRef]
  21. Heianna, J.; Makino, W.; Hirakawa, H.; Yamashita, Y.; Tomita, H.; Murayama, S. Therapeutic efficacy of intra-arterial docetaxel and nedaplatin infusion concomitant with radiotherapy for T4 maxillary sinus squamous cell carcinoma. Int. J. Oral. Maxillofac. Surg. 2022, 51, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  22. Yamashita, Y.; Ikegami, T.; Hirakawa, H.; Uehara, T.; Deng, Z.; Agena, S.; Uezato, J.; Kondo, S.; Kiyuna, A.; Maeda, H.; et al. Staging and prognosis of oropharyngeal carcinoma according to the 8th Edition of the American Joint Committee on Cancer Staging Manual in human papillomavirus infection. Eur. Arch. Otorhinolaryngol. 2019, 276, 827–836. [Google Scholar] [CrossRef]
  23. El-Mofty, S.K.; Lu, D.W. Prevalence of high-risk human papillomavirus DNA in nonkeratinizing (cylindrical cell) carcinoma of the sinonasal tract: A distinct clinicopathologic and molecular disease entity. Am. J. Surg. Pathol. 2005, 29, 1367–1372. [Google Scholar] [CrossRef] [PubMed]
  24. Bishop, J.A.; Guo, T.W.; Smith, D.F.; Wang, H.; Ogawa, T.; Pai, S.I.; Westra, W.H. Human papillomavirus-related carcinomas of the sinonasal tract. Am. J. Surg. Pathol. 2013, 37, 185–192. [Google Scholar] [CrossRef] [PubMed]
  25. Larque, A.B.; Hakim, S.; Ordi, J.; Nadal, A.; Diaz, A.; del Pino, M.; Marimon, L.; Alobid, I.; Cardesa, A.; Also, L. High-risk human papillomavirus is transcriptionally active in a subset of sinonasal squamous cell carcinomas. Mod. Pathol. 2014, 27, 343–351. [Google Scholar] [CrossRef]
  26. Chowdhury, N.; Alvi, S.; Kimura, K.; Tawfik, O.; Manna, P.; Beahm, D.; Robinson, A.; Kerley, S.; Hoover, L. Outcomes of HPV-related nasal squamous cell carcinoma. Laryngoscope 2017, 127, 1600–1603. [Google Scholar] [CrossRef]
  27. Also, L.; Moyano, S.; Nadal, A.; Alobid, I.; Blanch, J.L.; Ayala, E.; Lloveras, B.; Quint, W.; Cardesa, A.; Ordi, J. Human papillomaviruses are identified in a subgroup of sinonasal squamous cell carcinomas with favorable outcome. Cancer 2009, 115, 2701–2709. [Google Scholar] [CrossRef]
  28. Kılıç, S.; Kılıç, S.S.; Kim, E.S.; Baredes, S.; Mahmoud, O.; Gray, S.T.; Eloy, J.A. Significance of human papillomavirus positivity in sinonasal squamous cell carcinoma. Int. Forum. Allergy Rhinol. 2017, 7, 980–989. [Google Scholar] [CrossRef]
  29. Laco, J.; Sieglová, K.; Vošmiková, H.; Dundr, P.; Němejcová, K.; Michálek, J.; Čelakovský, P.; Chrobok, V.; Mottl, R.; Mottlová, A.; et al. The presence of high-risk human papillomavirus (HPV) E6/E7 mRNA transcripts in a subset of sinonasal carcinomas is evidence of involvement of HPV in its etiopathogenesis. Virchows Arch. 2015, 467, 405–415. [Google Scholar] [CrossRef]
  30. Nguyen, E.S.; Risbud, A.; Birkenbeuel, J.L.; Murphy, L.S.; Goshtasbi, K.; Pang, J.C.; Abiri, A.; Lehrich, B.M.; Haidar, Y.M.; Tjoa, T.; et al. Prognostic factors and outcomes of de novo sinonasal squamous cell carcinoma: A systematic review and meta-analysis. Otolaryngol. Head Neck Surg. 2022, 166, 434–443. [Google Scholar] [CrossRef]
  31. Suzuki, H.; Hanai, N.; Nishikawa, D.; Fukuda, Y.; Koide, Y.; Kodaira, T.; Tachibana, H.; Tomita, N.; Makita, C.; Hasegawa, Y. The Charlson comorbidity index is a prognostic factor in sinonasal tract squamous cell carcinoma. Jpn. J. Clin. Oncol. 2016, 46, 646–651. [Google Scholar] [CrossRef] [PubMed]
  32. Ma, S.J.; Yu, H.; Khan, M.; Yu, B.; Santhosh, S.; Chatterjee, U.; Gill, J.; Iovoli, A.; Farrugia, M.; Wooten, K.; et al. Defining the optimal threshold and prognostic utility of pre-treatment hemoglobin level as a biomarker for survival outcomes in head and neck cancer patients receiving chemoradiation. Oral. Oncol. 2022, 133, 106054. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, Y.; Cong, R.; Ji, C.; Ruan, W. The prognostic role of C-reactive protein in patients with head and neck squamous cell carcinoma: A meta-analysis. Cancer Med. 2020, 9, 9541–9553. [Google Scholar] [CrossRef] [PubMed]
  34. Dhanapal, R.; Saraswathi, T.; Govind, R.N. Cancer cachexia. J. Oral. Maxillofac. Pathol. 2011, 15, 257–260. [Google Scholar] [CrossRef] [PubMed]
  35. Doescher, J.; Piontek, G.; Wirth, M.; Bettstetter, M.; Schlegel, J.; Haller, B.; Brockhoff, G.; Reiter, R.; Pickhard, A. Epstein-Barr virus infection is strictly associated with the metastatic spread of sinonasal squamous-cell carcinomas. Oral. Oncol. 2015, 51, 929–934. [Google Scholar] [CrossRef]
  36. Sireci, F.; Dispenza, F.; Lorusso, F.; Immordino, A.; Immordino, P.; Gallina, S.; Peretti, G.; Canevari, F.R. Tumours of Nasal Septum: A Retrospective Study of 32 Patients. Int. J. Environ. Res. Public Health 2022, 19, 1713. [Google Scholar] [CrossRef]
  37. Megwalu, U.C.; Chen, M.M.; Ma, Y.; Divi, V. Surrogate for oropharyngeal cancer HPV status in cancer database studies. Head Neck 2017, 39, 2494–2500. [Google Scholar] [CrossRef]
  38. London, N.R., Jr.; Windon, M.J.; Amanian, A.; Zamuner, F.T.; Bishop, J.; Fakhry, C.; Rooper, L.M. Evaluation of the incidence of human papillomavirus-associated squamous cell carcinoma of the sinonasal tract among US adults. JAMA Netw. Open 2023, 6, e2255971. [Google Scholar] [CrossRef]
Figure 1. Selection of study participants. FFPE, formalin-fixed paraffin-embedded sample; n, number; SN, sinonasal.
Figure 1. Selection of study participants. FFPE, formalin-fixed paraffin-embedded sample; n, number; SN, sinonasal.
Jcm 12 06861 g001
Figure 2. Representative cases in p16 immunohistochemistry. p16 immunoreactivity was classified according to positive cell percentage and staining intensity of the tumor nucleus and cytosol ((A), none; (B), weak; (C), moderate; (D), strong). p16 overexpression was defined as having more than 75% of tumor cells positive with moderate to strong staining as in the representative cases.
Figure 2. Representative cases in p16 immunohistochemistry. p16 immunoreactivity was classified according to positive cell percentage and staining intensity of the tumor nucleus and cytosol ((A), none; (B), weak; (C), moderate; (D), strong). p16 overexpression was defined as having more than 75% of tumor cells positive with moderate to strong staining as in the representative cases.
Jcm 12 06861 g002
Figure 3. Representative findings for p16 overexpression in sinonasal squamous cell carcinoma. (A): hematoxylin–eosin staining. Bar = 50 µm. (B): p16 immunohistochemistry. Bar = 50 µm. (C): HPV DNA in situ hybridization. Bar = 50 µm. More than 75% of tumor cells expressed p16 immunoreactivity with moderate intensity and had a positive reaction in HPV DNA in situ hybridization.
Figure 3. Representative findings for p16 overexpression in sinonasal squamous cell carcinoma. (A): hematoxylin–eosin staining. Bar = 50 µm. (B): p16 immunohistochemistry. Bar = 50 µm. (C): HPV DNA in situ hybridization. Bar = 50 µm. More than 75% of tumor cells expressed p16 immunoreactivity with moderate intensity and had a positive reaction in HPV DNA in situ hybridization.
Jcm 12 06861 g003
Figure 4. Kaplan–Meier cumulative survival plots for OS according to T category (A), Hb level (B), and p16 overexpression status (C). The 5-year OS tended to be better in the p16-positive group (100%) than in the p16-negative group (71.2%) (p = 0.072). Hb, hemoglobin; OS, overall survival.
Figure 4. Kaplan–Meier cumulative survival plots for OS according to T category (A), Hb level (B), and p16 overexpression status (C). The 5-year OS tended to be better in the p16-positive group (100%) than in the p16-negative group (71.2%) (p = 0.072). Hb, hemoglobin; OS, overall survival.
Jcm 12 06861 g004
Figure 5. Kaplan–Meier cumulative survival plots for RFS according to the T category (A), ECOG PS (B), p16 overexpression (C), and mGPS (D). The five-year RFS rate was 69.9%. RFS in the p16-overexpression group was 90%, compared with 66.9% in the p16-negative group. However, the difference in RFS according to p16 overexpression was not significant in the univariate analysis (p = 0.142) but was significant in the multivariate analysis (p = 0.012). mGPS, modified Glasgow prognostic score; ECOG PS, Eastern Cooperative Oncology Group Performance Status; RFS, relapse-free survival.
Figure 5. Kaplan–Meier cumulative survival plots for RFS according to the T category (A), ECOG PS (B), p16 overexpression (C), and mGPS (D). The five-year RFS rate was 69.9%. RFS in the p16-overexpression group was 90%, compared with 66.9% in the p16-negative group. However, the difference in RFS according to p16 overexpression was not significant in the univariate analysis (p = 0.142) but was significant in the multivariate analysis (p = 0.012). mGPS, modified Glasgow prognostic score; ECOG PS, Eastern Cooperative Oncology Group Performance Status; RFS, relapse-free survival.
Jcm 12 06861 g005
Table 1. Clinical profiles in sinonasal squamous cell carcinoma patients.
Table 1. Clinical profiles in sinonasal squamous cell carcinoma patients.
VariablesTotal SNSCC (n = 79)HPV-Related
Tumor (n = 10)
HPV-Unrelated Tumor (n = 69)p-Value
Sex
Male707630.048
Female936
Median age 0.525
≤6139435
>6140634
Smoking history
Yes646580.070
No15411
Alcohol consumption
Yes678590.650
No12210
ECOG PS
0334290.903
1, 2, 346640
Primary subsite
Maxillary sinus63657
Ethmoid sinus1019
Nasal cavity633
Maxillary and ethmoid sinus736660.004
Nasal cavity633
Clinical T category
T1312
T2826
T322418
T4a31130
T4b15213
T1, T2, T3, T4a648560.930
T4b15213
Clinical N category
N052646
N11129
N215213
N3101
N0 526460.678
N1–327423
Clinical UICC stage
I312
II927
III13211
IVA, IVB54549
I, II, III235180.120
IV56551
Histology
Keratinizing75867* 0.076
Non-keratinizing422
Treatment 0.822
CRT/RT61853
Surgery ± RT/CRT18216
First medical examination * 0.037
Before 201535134
2015 or later44935
Prognosis
Alive without disease581048
Died of disease19019
Died of other cause202
CRT, chemoradiotherapy; ECOG PS, Eastern Cooperative Oncology Group Performance Status; RT, radiation therapy; SNSCC, sinonasal squamous cell carcinoma; UICC, 8th edition of the Union for International Cancer Control; * Fisher’s exact test; other categorical data were analyzed by chi-squared tests.
Table 2. Laboratory and nutritional variables in SNSCC.
Table 2. Laboratory and nutritional variables in SNSCC.
VariablesTotal SNSCC
(n = 79)
HPV-Related
Tumor (n = 10)
HPV-Unrelated
Tumor (n = 69)
p-Value
Hb (g/dL)
Median13.7
<13.7416350.583
≥13.738434
NLR (%)
Median3.19
<3.2405350.966
≥3.239534
Alb (g/dL)
Median3.9
<3.9374330.643
≥3.942636
CRP (mg/dL)
Median1.2
≤0.78376310.372
≥0.7842438
GNRI
<98313280.522
≥9848741
mGPS
A, B, C606540.207
D19415
PNI
<408260.268
≥4071863
Alb, albumin; CRP, C-reactive protein; GNRI, geriatric nutritional risk index; Hb, hemoglobin; mGPS, modified Glasgow prognostic score, NLR, neutrophil–lymphocyte rate; PNI, prognostic nutritional index; SNSCC, sinonasal squamous cell carcinoma.
Table 3. Distribution of p16 immunoreactivity.
Table 3. Distribution of p16 immunoreactivity.
Intensity ScoreNoneWeakModerateStrongTotal
% of positive cells04300043
1–200126018
20–5003508
50–7500000
≥75005510
Total431516579
Table 4. HPV analysis in p16 overexpression cases.
Table 4. HPV analysis in p16 overexpression cases.
Case SexAgeFirst Visit (Year)Primary SiteHPV TypeViral Load
(Copies/ng DNA)
E2/E6ISH
1M642014Nasal cavityHPV-5261,694.740.747Positive
2M612015MSNANANAPositive
3M662016MSNANANAPositive
4F462016ESHPV-182627.90Positive
5M652018Nasal cavityHPV-16141.60.0511Positive
6F632019MSNANANAPositive
7F532019Nasal cavityHPV-331.63 × 1060.8307Positive
8M802020MSNANANAPositive
9M462021MSHPV-164.30Positive
10M642021MSHPV-164.20Positive
ES, ethmoid sinus; F, female; ISH, in situ hybridization; M, male; MS, maxillary sinus; NA, not available.
Table 5. Univariate and multivariate analyses for OS and RFS.
Table 5. Univariate and multivariate analyses for OS and RFS.
OS RFS
Univariate
Analysis
Multivariate
Analysis
Univariate
Analysis
Multivariate
Analysis
Factor (Category)p-Valuep-ValueHR (95% CI)p-Valuep-ValueHR (95% CI)
Sex (male vs. female)0.335 0.547
Age (≤61 vs. ≥62 years)0.848 0.702
Tobacco history
(yes vs. no)
0.529 0.333
Alcohol consumption
(yes vs. no)
0.479 0.663
ECOG PS (0 vs. 1, 2, 3)0.273 0.0490.0260.364
(0.140–0.945)
Primary subsite
(nasal cavity vs. other)
0.652 0.516
Histology
(keratinizing vs. non-keratinizing)
0.921 0.815
p16 overexpression
or not
0.0720.004NA0.1420.0120.137
(0.018–1.073)
T category
(T1, T2, T3, T4a vs. T4b)
0.0110.0193.365
(1.312–8.630)
0.0010.0044.016
(1.68–9.61)
N category
(N0 vs. N, 1, 2, 3)
0.736 0.862
UICC clinical stage
(I, II, III vs. IV)
0.067 0.087
Hb (<13.7 vs. ≥13.7 g/dL)0.0120.0353.129
(1.016–9.6370)
0.0080.0742.458
(0.891–6.777)
NLR (≤3.2% vs. ≥3.2%)0.101 0.069
Alb (<3.9 vs. ≥3.9 g/dL)0.104 0.020
CRP (≥0.78 vs. <0.78 mg/dL)0.0580.6150.765
(0.265–2.210)
0.095
GNRI (<98 vs. ≥98)0.357 0.379
mGPS (A, B, C vs. D)0.166 0.0110.1870.540
(0.217–1.341)
PNI (<40 vs. ≥40)0.26 0.013
Treatment modality
(RT/CRT vs. surgery + RT/CRT)
0.575 0.836
Alb, albumin; CRP, C-reactive protein; CRT, chemoradiotherapy; ECOG PS, Eastern Cooperative Oncology Group Performance Status; GNRI, geriatric nutritional risk index; Hb, hemoglobin; HR, hazard ratio; mGPS, modified Glasgow prognostic score; NA, not available; NLR, neutrophil–lymphocyte rate; OS, overall survival; PNI, prognostic nutritional index; RFS, relapse-free survival; RT, radiation therapy; SNSCC, sinonasal squamous cell carcinoma; UICC, 8th edition of the Union for International Cancer Control.
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

Hirakawa, H.; Ikegami, T.; Touyama, M.; Ooshiro, Y.; Higa, T.; Higa, T.; Agena, S.; Kinjyo, H.; Kondo, S.; Kise, N.; et al. p16 Overexpression in Sinonasal Squamous Cell Carcinoma: Association with Human Papillomavirus and Prediction of Survival Outcomes. J. Clin. Med. 2023, 12, 6861. https://doi.org/10.3390/jcm12216861

AMA Style

Hirakawa H, Ikegami T, Touyama M, Ooshiro Y, Higa T, Higa T, Agena S, Kinjyo H, Kondo S, Kise N, et al. p16 Overexpression in Sinonasal Squamous Cell Carcinoma: Association with Human Papillomavirus and Prediction of Survival Outcomes. Journal of Clinical Medicine. 2023; 12(21):6861. https://doi.org/10.3390/jcm12216861

Chicago/Turabian Style

Hirakawa, Hitoshi, Taro Ikegami, Masatomo Touyama, Yurika Ooshiro, Tomoyo Higa, Teruyuki Higa, Shinya Agena, Hidetoshi Kinjyo, Shunsuke Kondo, Norimoto Kise, and et al. 2023. "p16 Overexpression in Sinonasal Squamous Cell Carcinoma: Association with Human Papillomavirus and Prediction of Survival Outcomes" Journal of Clinical Medicine 12, no. 21: 6861. https://doi.org/10.3390/jcm12216861

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