Skip to main content
Laryngoscope Investigative Otolaryngology logoLink to Laryngoscope Investigative Otolaryngology
. 2019 Oct 17;4(6):617–623. doi: 10.1002/lio2.316

A Hidden Epidemic of “Intermediate Risk” Oropharynx Cancer

Vlad C Sandulache 1,2,, David C Wilde 1, Erich M Sturgis 3, Elizabeth Y Chiao 4, Andrew G Sikora 1,2,
PMCID: PMC6929570  PMID: 31890879

Abstract

Objectives

Oropharyngeal squamous cell carcinoma (OPSCC) incidence is rapidly increasing in the United States and around the world, driven in large part by infection with the human papillomavirus (HPV). HPV associated OPSCC (HPV+OPSCC) has been shown to have improved response to treatment relative to tobacco‐associated OPSCC. However, improvement in patient survival has not been uniform. Subsets of OPSCC patients in the US and around the world continue to have poor oncologic outcomes. Although the drivers of this phenomenon remain unclear, there is increasing evidence that tobacco exposure plays an important role in modulating HPV+OPSCC clinical outcomes.

Methods

We conducted a review of the literature.

Results

We discuss the potential biological and epidemiological interplay between tobacco and HPV exposure in the context of OPSCC. Multiple retrospective and prospective cohorts show that HPV+OPSCC patients with a history of tobacco exposure have response to treatment and clinical outcomes distinct from HPV+OPSCC non‐smokers which poses clinical and scientific challenges to be addressed over the next decade.

Conclusions

The interaction between tobacco exposure and HPV infection in the context of OPSCC has significant implications for both standard of care treatment regimens and development of novel therapeutic approaches, in particular those which incorporate immunomodulatory agents.

Level of Evidence

5

Keywords: Oropharynx, HPV, tobacco, radiation, tumor immune microenvironment

RISING OROPHARYNGEAL SQUAMOUS CELL CARCINOMA INCIDENCE PRESENTS AN INCREASINGLY URGENT CLINICAL PROBLEM

Over the last two centuries, head and neck squamous cell carcinoma was a relatively rare entity, which was overwhelmingly attributable to tobacco and alcohol.1 In the United States, these cancers primarily occurred in older male smokers.1, 2 Oropharyngeal squamous cell carcinoma (OPSCC), a subset of head and neck squamous cell carcinomas, has been rising sharply in incidence over the last two decades with no evidence that this trend will soon abate.1, 3 This rise in incidence now affects most age groups, including those over the age of 65, a cohort traditionally associated with tobacco related OPSCC development.4 A recent analysis by Tota et al using registry data suggests a nearly 50% increase in the new cases of OPSCC diagnosed in the United States over the next two decades driven overwhelmingly by increased rates in elderly males.

Human papilloma virus (HPV) has long been known to be a major cause of cervical, penile, and anal cancer in the United States and the developing world.5, 6, 7 It is now clear that HPV is also the primary driver of the increase in OPSCC diagnoses. Over the last two decades, preclinical and clinical studies have conclusively linked HPV to OPSCC tumorigenesis in a majority of new diagnoses in the United States.8, 9, 10, 11, 12 Data from two Danish registry studies conducted over the first decade of this century demonstrate an incidence increase for HPV+OPSCC between 5% and 8% per year.13, 14 These data match that generated from a retrospective Dutch analysis which showed an increase in HPV positivity from 5% to nearly 30% over a two decade period.15 Similar changes in incidence have been demonstrated in several Asian studies (Korea: 2.4%16; Taiwan: 6.9%17). Together, these data indicate that HPV+OPSCC is becoming an urgent global clinical problem.

As discussed below, the effect of HPV on OPSCC incidence has fundamentally transformed the way we think about screening and prevention, the way we consider treatment escalation and/or de‐escalation, and the way we approach development of novel clinical trials.

NEW STAGING FOR HPV+OPSCC

In January of 2018, the 8th Edition of the AJCC Staging Manual was widely introduced into clinical practice in the United States. This change was prompted by a consistent finding of better survival for HPV+OPSCC as compared to HPV‐negative OPSCC in phase II and III clinical trials as well as large retrospective series showing that incorporation of HPV status into the staging system dramatically improved the risk stratification of OPSCC patients.1, 8, 18, 19 Consequently, a new staging system was created exclusively for HPV+OPSCC, and as a result of this change most patients previously staged as III–IV are now staged as I–II, reflecting their high disease‐free and overall survival (OS) at 2 years post‐treatment.

This improvement in HPV+OPSCC survival is both dramatic and unexpected. In contrast to OPSCC, carcinomas at most other head and neck sites have demonstrated either stagnate or only modestly improving survival rates over the last two decades.20, 21 Despite significant advancements in radiation and surgical techniques and the introduction of targeted agents (ie, tyrosine kinase inhibitors, monoclonal antibodies) into clinical practice throughout the United States, HPV remains the primary driver of improved survival recorded among head and neck sites.20, 21, 22, 23 In contrast to oral cavity and laryngeal SCC, OPSCC survival has nearly doubled over the last two decades, almost certainly driven by the epidemiological shift from HPV negative disease to a preponderance of HPV positive OPSCC.1, 24, 25

Although the improvement in clinical outcomes portends well for individual patients and the OPSCC patient population as whole, there exists significant data which suggest that not all HPV+OPSCC diagnoses can be expected to have excellent outcomes, and that much work remains to be done to better understand what drives HPV+OPSCC treatment response and patient survival. In the following sections, we discuss the modifying effect of tobacco exposure on HPV+OPSCC clinical outcomes.

NEW OPSCC DIAGNOSES OCCUR IN THE CONTEXT OF A PERSISTENTLY HIGH RATE OF TOBACCO EXPOSURE

Concomitant with an explosion in the incidence of HPV‐associated OPSCC (HPV+OPSCC), tobacco exposure, previously the primary carcinogenic driver of OPSCC has continually decreased in the US population.26 There are, however, important caveats to this general observation.

Our current understanding of HPV effects on OPSCC tumorigenesis includes a significant temporal latency of several decades between initial HPV exposure and development of HPV+OPSCC.27 As a result, for a majority of patients diagnosed today, initial HPV exposure is presumed to have occurred sometime between 20 and 30 years ago. For most individuals, smoking initiation occurs in the early teen years—20s; this is thus the primary formative time period for individual patients for deleterious behaviors such as continuation of tobacco exposure. Therefore, for patients with a new diagnosis of HPV+OPSCC, both HPV‐associated risk and tobacco exposure will be defined not by current rates, but by the epidemiology of HPV and smoking of the 1980s–2000s. According to the Centers for Disease Control and Prevention (CDC) in 1990, 50% of US adults reported being ever smokers, with the highest rate occurring among individuals age 25–44 and those with fewer than 12 years of education; 23% of individuals consumed more than 25 cigarettes per day.28 In 2000, the fraction of daily smokers remained at nearly 20%.29 By 2011, daily smoking was reported by only 15% of US adults and heavy smoking (>30 cigarettes/d) had decreased to <10% of the population.26

Reductions in smoking have not occurred at an even pace across all populations. Tobacco exposure in the US population ranges from 15% (non‐Hispanic whites) to 30% (Native American) of individuals26 and remains high among Veterans (40%–100%), elderly individuals, and individuals with low socioeconomic status.30, 31 In the US socioeconomic status and race impact not only smoking rates but also the ability of patients to quit, with non‐Hispanic blacks demonstrating the lowest rate and lowest interest in quitting which further decreased with advancing age. Recent increases in utilization of nontobacco‐based nicotine delivery products by teenagers and young adults raise the potential for future increases in tobacco exposure over the coming decades if a significant fraction of users ultimately crossover to tobacco containing products.32, 33 Reductions in smoking rates across the rest of the world have been equally slow and somewhat uneven across geographic regions and demographic strata.34, 35, 36

Based on these data, a significant fraction of new patients with a diagnosis of HPV+OPSCC both in the United States and around the world is expected to have a history of significant tobacco exposure and this will likely disproportionately impact minority patients with low socioeconomic status.

TOBACCO AND HPV LIKELY GENERATE A COMPLEX BIOLOGICAL INTERACTION IN OPSCC

Diseases, which are primarily associated with tobacco exposure, can occur in nonsmokers. In these instances, existing clinical and preclinical data point to disease variants with distinct biology and oncologic outcomes. For instance, lung cancers in nonsmokers (~25%) demonstrate such distinct behavior that these cases are now considered as a separate clinical entity.37, 38, 39, 40 OPSCC presents an even more complicated biological and oncologic dilemma. HPV‐associated OPSCC (HPV+OPSCC) is thought to be driven by inactivation of tumor suppressor pathways (ie, TP53, Rb) through direct and indirect interactions between viral proteins and host tumor cell proteins.41 In the context of tobacco‐associated squamous carcinomas in the head and neck region inactivating mutations in TP53 and other tumor suppressors are the primary drivers of tumor biology.42, 43 The publicly available HPV+OPSCC genomic and epigenetic data sets remain significantly limited with respect to size making a direct comparison challenging. The most recent the cancer genome atlas (TCGA) analysis completed by our group suggests differential methylation based on HPV status, with enrichment of the NANOG and MYC pathways but most importantly differential infiltration by favorable immunocyte populations.44 Unfortunately, this analysis was unable to more directly evaluate the interaction between tobacco and HPV status in a single disease site (OPSCC) due to limited sample size. The group from Memorial Sloan Kettering Cancer Center has completed the most direct head to head analysis of HPV+ and HPV− OPSCC and identified several potentially actionable differential deletions along with a higher frequency of TP53 mutations in HPV− OPSCC potentially related to tobacco exposure, yet a similar overall tumor mutation burden.45 These data are suggestive but far from definitive with respect to a direct molecular tobacco—HPV interaction at the level of tumor cells.

Approximately one in three new OPSCC has complex etiologic exposure signatures, where HPV and tobacco exposure overlap and contribute to tumorigenesis and treatment response in a manner which remains poorly understood to date. Ang et al demonstrated in the last decade that patients with HPV+OPSCC and a history of tobacco exposure formed an intermediate‐risk phenotype which manifested with worse survival compared to HPV+OPSCC in nonsmokers (<10 pack‐years) and improved survival compared to HPV‐OPSCC.8 This finding has since been confirmed in multiple retrospective and prospective data sets.46 It is important however to understand that the intermediate‐risk phenotype is described by clinical behavior not by a well‐understood biological mechanism. In large part, the difficulties associated with elucidating this interaction arise from two distinct but reinforcing problems. First, preclinical models which combine HPV and tobacco exposure are extremely limited and the basic science data are almost completely lacking. Second, clinical data sets provide very complex information which cannot easily help us focus our preclinical efforts as discussed in the following section.

TOBACCO UTILIZATION IS ASSOCIATED WITH DECREASED SURVIVAL IN HPV+OPSCC

Multiple clinical data sets indicate that tobacco exposure modulates clinical outcomes in HPV+OPSCC.8 Most recently, Vawda et al found lower relapse‐free survival and OS in current smokers versus never smokers or former smokers and a relative dose dependent decrease in OS as tobacco exposure increased (pack‐year history). The relative effect size for OS exceeded 20%, a striking clinical effect size within the context of OPSCC equal to or greater than the impact of positive nodal status or extranodal extension and more than double the effect size of adding cisplatin to radiation‐based treatment.21, 47, 48, 49 These data are consistent with a reanalysis of RTOG 0129 and RTOG 9003 data showing that risk of disease progression or death increased with increasing tobacco consumption as well as retrospective single institution series which demonstrated higher rates of recurrence and decreased survival among HPV+OPSCC patients with a history of tobacco exposure.50, 51 Precisely how this effect is generated however remains unclear.

HPV clearly contributes to the development of OPSCC in the presence or absence of tobacco exposure in a manner which provides epidemiological support for individual‐to‐individual transmission, through sexual encounters.52 Conversely, HPV− OPSCC is associated with tobacco and alcohol exposure but not behavioral patterns related to sexual activity.9 Whether alcohol or tobacco use increase risk of HPV+OPSCC remains debatable.53 There are some data sets, which do suggest that HPV and tobacco exposure, may be synergistic in terms of increasing risk. In women, smoking history has been found to impact disease progression to HPV‐mediated cancer.54 Kreimer et al showed in a multinational cohort of healthy men a relatively low frequency of high‐risk HPV strains which was relatively consistent across countries, but was increased 2.5‐fold in the context of current tobacco use.55 Anantharaman et al examined the interaction between HPV and smoking in head and neck cancer patients and control participants from multiple European centers and identified an additive effect on OPSCC development.56 Smith et al concluded that tobacco and alcohol use impacted the relative risk of cancer development regardless of HPV serologic status.57

In addition to direct effects on OPSCC tumorigenesis, tobacco exposure is a critical driver of overall patient health. This includes a dramatic reduction in pulmonary health and an increased rate of chronic pulmonary disease58, 59 as well as an increased risk of cardiovascular disease and fatal cardiac events.60, 61, 62 A large prospective study of British physicians identified a 2‐fold increase in all‐cause mortality associated with smoking, driven in large part by development of tobacco‐associated malignancies and development of cardiopulmonary disease.63 In the US population, smoking has been associated with a 3‐fold higher rate of death and a reduction in life expectancy of over one decade.64 Together, these data suggest that HPV+OPSCC smokers will have decreased long‐term survival in part as a result of tobacco mediated effects on overall health and comorbidity burden. In addition, tobacco‐associated comorbidities are also likely to impact short‐term, disease specific survival through decreased treatment tolerance. Multiple studies have shown that patients with a higher comorbidity burden experience greater rates of treatment de‐escalation and/or cessation/interruption resulting in reduced treatment efficacy and disease control.65, 66

In summary, tobacco can interact directly with HPV as it relates to tumor biology in addition to modulating the overall health/all‐cause mortality of any cohort of smokers with HPV+OPSCC. Since frail patients with a higher comorbidity burden will also generally demonstrate decreased treatment tolerance and more frequent treatment de‐intensification, it is possible that the interaction between HPV and tobacco as it relates to clinical outcomes in OPSCC is in fact driven by at least three overlapping clinical/epidemiological phenomena which cannot be easily dissected using retrospective analysis of clinical data sets.

VARIABLE CLINICAL OUTCOMES FOR SUBSETS OF OPSCC PATIENTS

The staging change for HPV+OPSCC was foreshadowed by a comprehensive analysis from the University of Texas MD Anderson Cancer Center which demonstrated a clear progression in clinical outcomes as a function of disease characteristics. Specifically, younger patients, without a history of tobacco exposure and early T‐classification tumors, demonstrated a significant improvement in survival post‐1995 compared to the previous half century. This transition was so dramatic that it erased the normal correlation between tumor, nodal and metastasis classification and survival, primarily due to the biological and treatment effects of HPV.1 However, that same study as well as the Ang et al data set identified a subset of OPSCC patients with poor disease‐free survival and OS, in line with historical data.1, 8

More recently, analyses of subsets of US patients have indicated that the expected improvement in HPV+OPSCC survival is far from uniform. Several analyses of Veterans showed that a majority of patients maintained a high rate of heavy tobacco exposure.2, 67 Despite a positive impact of HPV positivity on disease‐free survival and OS, survival rates for both HPV+ and HPV− disease were in line with the Ang et al intermediate‐risk and high‐risk rates and the low‐risk group was essentially absent.2, 68 These characteristics were conserved in both White and African American patients, resulting in similar disease behavior and oncologic outcomes.69 Data from RTOG 0129 and RTOG 0522 have been reanalyzed with a longer follow‐up period and demonstrated that the OS and progression‐free survival (PFS) rates for low‐, intermediate‐, and high‐risk OPSCC patients persistent with a difference in PFS between low‐ and intermediate‐risk groups of over 15%.70

Together these data provide a sobering reminder that not all new HPV+OPSCC patients should be expected to demonstrate uniform clinical outcomes and excellent survival. This is critically important in light of recent attempts to tailor treatment regimens to OPSCC to maximize survival and reduce treatment related toxicity.

TREATMENT ALGORITHMS AND CHANGING PARADIGMS

Treatment paradigms of the 1990s and 2000s were focused on maintaining therapeutic equipoise and escalation. These efforts are perhaps best exemplified by the PARADIGM and DeCIDE clinical trials which tested the addition of induction chemotherapy to concurrent chemo‐radiation to improve survival rates.71, 72 Neither study demonstrated an improvement in survival through the addition of induction chemotherapy although survival rates for both the standard of care and experimental arms were substantially higher than would have been expected from previous data sets and accrual was incomplete. Although failure to complete accrual was partially blamed for the failure of these studies, it is quite possible that these studies failed to detect an improvement in survival because outcomes were already improving in patients with advanced‐stage OPSCC.

Current therapeutic paradigms have shifted in light of improved survival data for HPV+OPSCC. Given the excellent survival of most HPV+OPSCC patients, there is now an appropriate focus on de‐escalation of treatment intensity to ameliorate treatment‐related toxicity. This is particularly important as the demographics of HPV+OPSCC shift to an older patient cohort with more expected comorbidities and potentially lower tolerance for intense treatment regimens.73 The first large randomized study to tackle the approach of de‐escalation, RTOG 1016 tested whether cetuximab would be able to deliver noninferior OS and PFS compared to a cisplatin‐based regimen. The trial focused exclusively on patients with HPV+OPSCC generating a homogeneity of patient cohort not present in any previous prospective clinical trial of this size. Despite utilization of an accelerated fractionation regimen, both OS and PFS were significantly inferior in the cetuximab arm of the trial compared to the cisplatin arm with a difference of over 10% in PFS at 5 years.24 By extrapolation, it is possible that in a patient cohort enriched for intermediate‐risk OPSCC, the inferiority of cetuximab could potentially have been even more dramatic.

Multiple other studies have been initiated in the last decade with a focus on de‐escalation using a variety of approaches, including incorporation of surgery, de‐escalation of radiation dose, and changes in chemotherapy strategies. The advent of immunotherapy and its introduction into the clinical armamentarium of oncologists treating OPSCC has added yet another dimension to these ongoing efforts at precision oncology for HPV+OPSCC. CheckMate 141 generated promising results, with nivolumab demonstrating a significant increase in OS compared to nonimmunomodulatory systemic therapy and importantly a more favorable toxicity profile. These effects were measured in both p16+ and p16− tumors although the benefit appeared more pronounced in p16+ tumors.74, 75 It is expected that introduction of immunomodulatory agents earlier in the treatment course could generate even more favorable effects, although this remains to be demonstrated. Similarly, whether immunomodulatory agents can improve oncologic outcomes while decreasing overall treatment‐related toxicity in low‐ and intermediate‐risk OPSCC remains an open question.

As pointed out by multiple investigators, however, it is critical that current trials aimed at de‐escalation regardless of approach (ie, targeted agents, immunotherapy) consider very carefully criteria for inclusion of patients.70 In our opinion, despite difficulties associated with accurately ascertaining and quantifying tobacco exposure, significant efforts should be dedicated to the addition of tobacco exposure into inclusion criteria for de‐intensification trials. As discussed below, this consideration is potentially even more important when considering incorporation of targeted agents and immunomodulatory agents into novel treatment regimens.

VARIABLE RESPONSE RATES SUGGEST DIFFERENTIAL BIOLOGY WITHIN THE SPECTRUM OF HPV+OPSCC

Despite the clear separation with regard to treatment response and survival generated by HPV status, there is increasingly clear evidence that HPV+OPSCC tumors do not behave in a homogeneous manner. HPV+OPSCC demonstrates unique clinical behaviors such as distant metastases to bone and solid organs other than lung.11 This pattern of metastasis occurs over a delayed time frame and has the potential to radically change the manner in which surveillance for recurrence/distant metastasis is performed in this patient population.76 Specifically, distant metastases to solid organs including brain will require some modification of normal clinical surveillance questioning related to patient symptoms and also likely increase the relative utility of whole body positron emission tomography and dedicated brain imaging over the next 1–2 decades.77, 78, 79

Within the spectrum of HPV+OPSCC, gene expression profiles partially related to tumor metabolism have been recently shown to predict stratification of clinical outcomes and may serve as a future biomarker for treatment selection.10 Precisely what drives stratification of biological behavior and treatment response in HPV+OPSCC remains unclear. However, we believe that data from other tumors strongly support a role for tobacco exposure. Castelletti et al showed distinct molecular models for lung cancer patients with a history of radiation exposure compared to patients with a history of tobacco exposure.80 More specifically, tobacco exposure has been shown to impact patterns of KRAS mutations in lung cancers.81, 82 In addition, smoking has been shown to have a significant impact on genome wide methylation which can persist even following cessation of smoking.83 Although mutations and methylation are likely to demonstrate long‐term persistence, smoking‐related changes in gene expression do appear to reverse following cessation, although the relative timing remains both unclear and highly variable.84 Interestingly, the recent analysis by Harbison et al of HPV+OPSCC tumor with documented recurrence identified a mutational landscape similar to HPV‐unrelated HNSCC tumors and metachronous recurrent OPSCC demonstrated a profile very similar to traditional, tobacco related tumors, including TP53, CASP8, and FAT1 mutational events.85 Although too small to address the question of tobacco exposure as a driver of the intermediate‐risk phenotype and potentially genotype, this study does support the hypothesized biological interaction between conventional carcinogen exposure and HPV in the context of OPSCC.

In addition to driving tumor (parasite) biology, tobacco exposure also exhibits significant effects on host biology through alterations of antitumor and systemic immunity and inflammation.86 Tobacco effects on circulating immunocytes have been well established in the literature and have been shown to be both profound and relatively cell type specific.87, 88, 89 Most recently, analysis of TCGA data for both head and neck squamous cell carcinoma and lung squamous cell carcinoma demonstrated a smoking signature that includes a higher mutational load and variable effects on tumor immunity. Critically, smoking generated opposing effects in head and neck squamous cell carcinoma and lung squamous cell carcinoma as it relates to tumor immunity, highlighting our difficulties in generating rational immunomodulatory approaches for HPV+OPSCC.90 This difficulty is further augmented by the clear interaction between tumor characteristics (ie, mutational burden/profile) and the tumor immune microenvironment, generating differential responses to immunomodulatory agents as has been demonstrated in lung cancer patients.91

CONCLUSIONS

HPV+OPSCC is an increasingly prevalent clinical problem in the United States and around the world. Fortunately, many HPV+OPSCC patients have an excellent response to treatment, resulting in concomitant excellent survival. This has prompted significant changes in staging, and increased interest in de‐escalation of treatment intensity. However, it is now clear that within the spectrum of HPV+OPSCC disease, there are patient subsets which continue to experience poor treatment response and survivorship. These patients have a disproportionate exposure to tobacco, which may explain their different survival characteristics. Although the interaction between HPV and tobacco in the context of OPSCC remains poorly understood and is likely multifactorial, its effects on clinical outcomes are important and cannot be ignored. Focused investigation is needed to elucidate this critical intersection of biology and epidemiology to better understand the behavior and optimize the treatment of intermediate‐risk OPSCC.

It is important to note that even current efforts aimed directly at improving outcomes for intermediate‐risk HPV+OPSCC such as EA3161 (phase II/III Randomized Study of Maintenance Nivolumab vs. Observation in Patients with Locally Advanced, Intermediate Risk HPV‐Positive Oropharyngeal Cancer) are essentially based solely on a risk‐stratification schema defined by reported tobacco exposure as opposed to clear biological framework for this disease. In some ways, this parallels the experience with de‐escalation regimens targeting HPV+OPSCC more broadly since the mechanisms which might drive increased chemo‐ and radio‐sensitivity in HPV driven tumors continue to remain unclear. In our opinion, there is a critical need for well‐defined translational efforts in this area using preclinical models of HPV+OPSCC and preclinical models of acute and chronic tobacco exposure which can allow us to study both intrinsic tumor biology and the tumor immune microenvironment associated with HPV+OPSCC. Without such preclinical models, mechanistic studies will continue to lag clinical data, and we risk failure of multiple current and planned therapeutic clinical trials.

Editor's Note: This Manuscript was accepted for publication on September 21, 2019.

Conflict of Interest: The authors have no conflicts of interest to disclose related to the material included in this article.

Funding: There are no external funding sources related to the material included in this article.

Contributor Information

Vlad C. Sandulache, Email: vlad.sandulache@bcm.edu.

Andrew G. Sikora, Email: andrew.sikora@bcm.edu.

BIBLIOGRAPHY

  • 1. Dahlstrom KR, Calzada G, Hanby JD, et al. An evolution in demographics, treatment, and outcomes of oropharyngeal cancer at a major cancer center: a staging system in need of repair. Cancer 2013;119:81–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Sandulache VC, Hamblin J, Lai S, et al. Oropharyngeal squamous cell carcinoma in the veteran population: association with traditional carcinogen exposure and poor clinical outcomes. Head Neck 2015;37:1246–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Chaturvedi AK, Engels EA, Pfeiffer RM, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol 2011;29:4294–4301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Lu DJ, Luu M, Mita A, et al. Human papillomavirus‐associated oropharyngeal cancer among patients aged 70 and older: dramatically increased prevalence and clinical implications. Eur J Cancer 2018;103:195–204. [DOI] [PubMed] [Google Scholar]
  • 5. Gupta SM, Mania‐Pramanik J. Molecular mechanisms in progression of HPV‐associated cervical carcinogenesis. J Biomed Sci 2019;26:28. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 6. Castellsague X. Natural history and epidemiology of HPV infection and cervical cancer. Gynecol Oncol 2008;110:S4–S7. [DOI] [PubMed] [Google Scholar]
  • 7. Shrestha AD, Neupane D, Vedsted P, Kallestrup P. Cervical cancer prevalence, incidence and mortality in low and middle income countries: a systematic review. Asian Pac J Cancer Prev 2018;19:319–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 2010;363:24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gillison ML, D'Souza G, Westra W, et al. Distinct risk factor profiles for human papillomavirus type 16‐positive and human papillomavirus type 16‐negative head and neck cancers. J Natl Cancer Inst 2008;100:407–420. [DOI] [PubMed] [Google Scholar]
  • 10. Gleber‐Netto FO, Rao X, Guo T, et al. Variations in HPV function are associated with survival in squamous cell carcinoma. JCI Insight 2019;4(1):124762. 10.1172/jci.insight.124762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Muller S, Khuri FR, Kono SA, Beitler JJ, Shin DM, Saba NF. HPV positive squamous cell carcinoma of the oropharynx. Are we observing an unusual pattern of metastases? Head Neck Pathol 2012;6:336–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Shay SG, Chang E, Lewis MS, Wang MB. Characteristics of human papillomavirus‐associated head and neck cancers in a veteran population. JAMA Otolaryngol Head Neck Surg 2015;141:790–796. [DOI] [PubMed] [Google Scholar]
  • 13. Garnaes E, Kiss K, Andersen L, et al. A high and increasing HPV prevalence in tonsillar cancers in Eastern Denmark, 2000–2010: the largest registry‐based study to date. Int J Cancer 2015;136:2196–2203. [DOI] [PubMed] [Google Scholar]
  • 14. Garnaes E, Kiss K, Andersen L, et al. Increasing incidence of base of tongue cancers from 2000 to 2010 due to HPV: the largest demographic study of 210 Danish patients. Br J Cancer 2015;113:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Rietbergen MM, Leemans CR, Bloemena E, et al. Increasing prevalence rates of HPV attributable oropharyngeal squamous cell carcinomas in the Netherlands as assessed by a validated test algorithm. Int J Cancer 2013;132:1565–1571. [DOI] [PubMed] [Google Scholar]
  • 16. Shin A, Jung YS, Jung KW, Kim K, Ryu J, Won YJ. Trends of human papillomavirus‐related head and neck cancers in Korea: national cancer registry data. Laryngoscope 2013;123:E30–E37. [DOI] [PubMed] [Google Scholar]
  • 17. Hwang T‐Z, Hsiao J‐R, Tsai C‐R, Chang JS. Incidence trends of human papillomavirus‐related head and neck cancer in Taiwan, 1995–2009. Int J Cancer 2015;137:395–408. [DOI] [PubMed] [Google Scholar]
  • 18. Lydiatt WM, Patel SG, O'Sullivan B, et al. Head and neck cancers‐major changes in the American Joint Committee on cancer eighth edition cancer staging manual. CA Cancer J Clin 2017;67:122–137. [DOI] [PubMed] [Google Scholar]
  • 19. O'Sullivan B, Huang SH, Su J, et al. Development and validation of a staging system for HPV‐related oropharyngeal cancer by the International Collaboration on Oropharyngeal Cancer Network for Staging (ICON‐S): a multicentre cohort study. Lancet Oncol 2016;17:440–451. [DOI] [PubMed] [Google Scholar]
  • 20. Hoffman HT, Porter K, Karnell LH, et al. Laryngeal cancer in the United States: changes in demographics, patterns of care, and survival. Laryngoscope 2006;116:1–13. [DOI] [PubMed] [Google Scholar]
  • 21. Sandulache VC, Michikawa C, Kataria P, et al. High‐risk TP53 mutations are associated with extranodal extension in oral cavity squamous cell carcinoma. Clin Cancer Res 2018;24:1727–1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Bonner J, Giralt J, Harari P, et al. Cetuximab and radiotherapy in laryngeal preservation for cancers of the larynx and hypopharynx: a secondary analysis of a randomized clinical trial. JAMA Otolaryngol Head Neck Surg 2016;142:842–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for squamous‐cell carcinoma of the head and neck. N Engl J Med 2006;354:567–578. [DOI] [PubMed] [Google Scholar]
  • 24. Gillison ML, Trotti AM, Harris J, et al. Radiotherapy plus cetuximab or cisplatin in human papillomavirus‐positive oropharyngeal cancer (NRG Oncology RTOG 1016): a randomised, multicentre, non‐inferiority trial. Lancet 2019;393:40–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Mehanna H, Robinson M, Hartley A, et al. Radiotherapy plus cisplatin or cetuximab in low‐risk human papillomavirus‐positive oropharyngeal cancer (De‐ESCALaTE HPV): an open‐label randomised controlled phase 3 trial. Lancet 2019;393:51–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Agaku IT, King BA, Dube SR, Centers for Disease Control and Prevention . Current cigarette smoking among adults ‐ United States, 2005–2012. MMWR Morb Mortal Wkly Rep 2014;63:29–34. [PMC free article] [PubMed] [Google Scholar]
  • 27. Gravitt PE, Winer RL. Natural history of HPV infection across the lifespan: role of viral latency. Viruses 2017;9(10):E267. 10.3390/v9100267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Centers for Disease Control (CDC) . Cigarette smoking among adults—United States, 1990. MMWR Morb Mortal Wkly Rep 1992;41:354–355. 361–362. [PubMed] [Google Scholar]
  • 29. Trosclair A, Husten C, Pederson L, Dhillon I. Cigarette smoking among adults—United States, 2000. MMWR Morb Mortal Wkly Rep 2002;51:642–645. [PubMed] [Google Scholar]
  • 30. Klevens RM, Giovino GA, Peddicord JP, Mowery P, Grummer‐Strawn L, Nelson DE. The association between veteran status and cigarette‐smoking behaviors. Am J Prev Med 1995;11:245–250. [PubMed] [Google Scholar]
  • 31. Wang TW, Asman K, Gentzke AS, Cullen KA, Holder‐Hayes E. Tobacco product use among adults—United States, 2017. MMWR Morb Mortal Wkly Rep 2018;67:1225–1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Leventhal AM, Strong DR, Kirkpatrick MG, et al. Association of electronic cigarette use with initiation of combustible tobacco product smoking in early adolescenceassociation of e‐cigarette use with smoking during early adolescenceassociation of e‐cigarette use with smoking during early adolescence. JAMA 2015;314:700–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Coleman BN, Apelberg BJ, Ambrose BK, et al. Association between electronic cigarette use and openness to cigarette smoking among US young adults. Nicotine Tob Res 2015;17:212–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Han J, Chen X. A meta‐analysis of cigarette smoking prevalence among adolescents in China: 1981‐2010. Int J Environ Res Public Health 2015;12:4617–4630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Stefler D, Azarova A, Irdam D, et al. Smoking, alcohol and cancer mortality in Eastern European men: findings from the PrivMort retrospective cohort study. Int J Cancer 2018;143:1128–1133. [DOI] [PubMed] [Google Scholar]
  • 36. GBD 2017 Risk Factor Collaborators . Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990‐2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018;392:1923–1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Chapman AM, Sun KY, Ruestow P, Cowan DM, Madl AK. Lung cancer mutation profile of EGFR, ALK, and KRAS: meta‐analysis and comparison of never and ever smokers. Lung Cancer 2016;102:122–134. [DOI] [PubMed] [Google Scholar]
  • 38. Huang Y, Wang R, Pan Y, et al. Clinical and genetic features of lung squamous cell cancer in never‐smokers. Oncotarget 2016;7:35979–35988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Li Y, Sheu CC, Ye Y, et al. Genetic variants and risk of lung cancer in never smokers: a genome‐wide association study. Lancet Oncol 2010;11:321–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers—a different disease. Nat Rev Cancer 2007;7:778–790. [DOI] [PubMed] [Google Scholar]
  • 41. Rampias T, Sasaki C, Psyrri A. Molecular mechanisms of HPV induced carcinogenesis in head and neck. Oral Oncol 2014;50:356–363. [DOI] [PubMed] [Google Scholar]
  • 42. Agrawal N, Frederick MJ, Pickering CR, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 2011;333:1154–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Cancer Genome Atlas Network . Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015;517:576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Carrero I, Liu HC, Sikora AG, Milosavljevic A. Histoepigenetic analysis of HPV‐ and tobacco‐associated head and neck cancer identifies both subtype‐specific and common therapeutic targets despite divergent microenvironments. Oncogene 2019;38:3551–3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Dogan S, Xu B, Middha S, et al. Identification of prognostic molecular biomarkers in 157 HPV‐positive and HPV‐negative squamous cell carcinomas of the oropharynx. Int J Cancer 2019. 10.1002/ijc.32412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Fakhry C, Zhang Q, Nguyen‐Tan PF, et al. Human papillomavirus and overall survival after progression of oropharyngeal squamous cell carcinoma. J Clin Oncol 2014;32:3365–3373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Vawda N, Banerjee RN, Debenham BJ. Impact of smoking on outcomes of HPV‐related oropharyngeal cancer treated with primary radiation or surgery. Int J Radiat Oncol Biol Phys 2019;103:1125–1131. [DOI] [PubMed] [Google Scholar]
  • 48. Pignon JP, le Maitre A, Maillard E, Bourhis J, MACH‐NC Collaborative Group . Meta‐analysis of chemotherapy in head and neck cancer (MACH‐NC): an update on 93 randomised trials and 17,346 patients. Radiother Oncol 2009;92:4–14. [DOI] [PubMed] [Google Scholar]
  • 49. Blanchard P, Baujat B, Holostenco V, et al. Meta‐analysis of chemotherapy in head and neck cancer (MACH‐NC): a comprehensive analysis by tumour site. Radiother Oncol 2011;100:33–40. [DOI] [PubMed] [Google Scholar]
  • 50. Gillison ML, Zhang Q, Jordan R, et al. Tobacco smoking and increased risk of death and progression for patients with p16‐positive and p16‐negative oropharyngeal cancer. J Clin Oncol 2012;30:2102–2111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Maxwell JH, Kumar B, Feng FY, et al. Tobacco use in human papillomavirus‐positive advanced oropharynx cancer patients related to increased risk of distant metastases and tumor recurrence. Clin Cancer Res 2010;16:1226–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. D'Souza G, Kreimer AR, Viscidi R, et al. Case‐control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 2007;356:1944–1956. [DOI] [PubMed] [Google Scholar]
  • 53. Applebaum KM, Furniss CS, Zeka A, et al. Lack of association of alcohol and tobacco with HPV16‐associated head and neck cancer. J Natl Cancer Inst 2007;99:1801–1810. [DOI] [PubMed] [Google Scholar]
  • 54. Castellsagué X, Bosch FX, Muñoz N. Environmental co‐factors in HPV carcinogenesis. Virus Res 2002;89:191–199. [DOI] [PubMed] [Google Scholar]
  • 55. Kreimer AR, Villa A, Nyitray AG, et al. The epidemiology of oral HPV infection among a multinational sample of healthy men. Cancer Epidemiol Biomarkers Prev 2011;20:172–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Anantharaman D, Muller DC, Lagiou P, et al. Combined effects of smoking and HPV16 in oropharyngeal cancer. Int J Epidemiol 2016;45:752–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Smith EM, Rubenstein LM, Haugen TH, Hamsikova E, Turek LP. Tobacco and alcohol use increases the risk of both HPV‐associated and HPV‐independent head and neck cancers. Cancer Causes Control 2010;21:1369–1378. [DOI] [PubMed] [Google Scholar]
  • 58. Løkke A, Lange P, Scharling H, Fabricius P, Vestbo J. Developing COPD: a 25 year follow up study of the general population. Thorax 2006;61:935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Pelkonen M, Notkola I‐L, Nissinen A, Tukiainen H, Koskela H. Thirty‐year cumulative incidence of chronic bronchitis and COPD in relation to 30‐year pulmonary function and 40‐year mortality: a follow‐up in middle‐aged rural men. Chest 2006;130:1129–1137. [DOI] [PubMed] [Google Scholar]
  • 60. Willett WC, Green A, Stampfer MJ, et al. Relative and absolute excess risks of coronary heart disease among women who smoke cigarettes. N Engl J Med 1987;317:1303–1309. [DOI] [PubMed] [Google Scholar]
  • 61. Bjartveit K, Tverdal A. Health consequences of smoking 1–4 cigarettes per day. Tob Control 2005;14:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Blanco‐Cedres L, Daviglus ML, Garside DB, et al. Relation of cigarette smoking to 25‐year mortality in middle‐aged men with low baseline serum cholesterol : the Chicago Heart Association Detection Project in industry. Am J Epidemiol 2002;155:354–360. [DOI] [PubMed] [Google Scholar]
  • 63. Doll R, Peto R, Wheatley K, Gray R, Sutherland I. Mortality in relation to smoking: 40 years' observations on male British doctors. BMJ 1994;309:901–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Jha P, Ramasundarahettige C, Landsman V, et al. 21st‐century hazards of smoking and benefits of cessation in the United States. N Engl J Med 2013;368:341–350. [DOI] [PubMed] [Google Scholar]
  • 65. Fesinmeyer MD, Mehta V, Tock L, Blough D, McDermott C, Ramsey SD. Completion of radiotherapy for local and regional head and neck cancer in medicare. JAMA Otolaryngol Head Neck Surg 2009;135:860–867. [DOI] [PubMed] [Google Scholar]
  • 66. Lazarev S, Gupta V, Ghiassi‐Nejad Z, et al. Premature discontinuation of curative radiation therapy: insights from head and neck irradiation. Adv Radiat Oncol 2017;3:62–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Park J, McPike V, Kambhampati S, et al. Positivity rates in oropharyngeal and nonoropharyngeal head and neck cancer in the VA. Fed Pract 2018;35:S44–S47. [PMC free article] [PubMed] [Google Scholar]
  • 68. Feinstein AJ, Shay SG, Chang E, Lewis MS, Wang MB. Treatment outcomes in veterans with HPV‐positive head and neck cancer. Am J Otolaryngol 2017;38:188–192. [DOI] [PubMed] [Google Scholar]
  • 69. Zevallos JP, Sandulache VC, Hamblin J, et al. Impact of race on oropharyngeal squamous cell carcinoma presentation and outcomes among veterans. Head Neck 2016;38:44–50. [DOI] [PubMed] [Google Scholar]
  • 70. Fakhry C, Zhang Q, Gillison ML, et al. Validation of NRG oncology/RTOG‐0129 risk groups for HPV‐positive and HPV‐negative oropharyngeal squamous cell cancer: implications for risk‐based therapeutic intensity trials. Cancer 2019;125(12):2027–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Haddad R, O'Neill A, Rabinowits G, et al. Induction chemotherapy followed by concurrent chemoradiotherapy (sequential chemoradiotherapy) versus concurrent chemoradiotherapy alone in locally advanced head and neck cancer (PARADIGM): a randomised phase 3 trial. Lancet Oncol 2013;14:257–264. [DOI] [PubMed] [Google Scholar]
  • 72. Cohen EE, Karrison TG, Kocherginsky M, et al. Phase III randomized trial of induction chemotherapy in patients with N2 or N3 locally advanced head and neck cancer. J Clin Oncol 2014;32:2735–2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Tota JE, Best AF, Zumsteg ZS, Gillison ML, Rosenberg PS, Chaturvedi AK. Evolution of the oropharynx cancer epidemic in the United States: moderation of increasing incidence in younger individuals and shift in the burden to older individuals. J Clin Oncol 2019;37:1538–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab vs investigator's choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2‐year long‐term survival update of CheckMate 141 with analyses by tumor PD‐L1 expression. Oral Oncol 2018;81:45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for recurrent squamous‐cell carcinoma of the head and neck. N Engl J Med 2016;375:1856–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Trosman SJ, Koyfman SA, Ward MC, et al. Effect of human papillomavirus on patterns of distant metastatic failure in oropharyngeal squamous cell carcinoma treated with chemoradiotherapy. JAMA Otolaryngol Head Neck Surg 2015;141:457–462. [DOI] [PubMed] [Google Scholar]
  • 77. Huang SH, Perez‐Ordonez B, Liu F‐F, et al. Atypical clinical behavior of p16‐confirmed HPV‐related oropharyngeal squamous cell carcinoma treated with radical radiotherapy. Int J Radiat Oncol Biol Phys 2012;82:276–283. [DOI] [PubMed] [Google Scholar]
  • 78. Huang SH, Perez‐Ordonez B, Weinreb I, et al. Natural course of distant metastases following radiotherapy or chemoradiotherapy in HPV‐related oropharyngeal cancer. Oral Oncol 2013;49:79–85. [DOI] [PubMed] [Google Scholar]
  • 79. Bulut OC, Lindel K, Hauswald H, et al. Clinical and molecular characteristics of HNSCC patients with brain metastases: a retrospective study. Eur Arch Otorhinolaryngol 2014;271:1715–1722. [DOI] [PubMed] [Google Scholar]
  • 80. Castelletti N, Kaiser JC, Simonetto C, Furukawa K, Kuchenhoff H, Stathopoulos GT. Risk of lung adenocarcinoma from smoking and radiation arises in distinct molecular pathways. Carcinogenesis 2019:bgz036. 10.1093/carcin/bgz036. [DOI] [PubMed] [Google Scholar]
  • 81. Kim HR, Ahn JR, Lee JG, et al. The impact of cigarette smoking on the frequency of and qualitative differences in KRAS mutations in Korean patients with lung adenocarcinoma. Yonsei Med J 2013;54:865–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Riely GJ, Kris MG, Rosenbaum D, et al. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res 2008;14:5731–5734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Joehanes R, Just AC, Marioni RE, et al. Epigenetic signatures of cigarette smoking. Circ Cardiovasc Genet 2016;9:436–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bosse Y, Postma DS, Sin DD, et al. Molecular signature of smoking in human lung tissues. Cancer Res 2012;72:3753–3763. [DOI] [PubMed] [Google Scholar]
  • 85. Harbison RA, Kubik M, Konnick EQ, et al. The mutational landscape of recurrent versus nonrecurrent human papillomavirus‐related oropharyngeal cancer. JCI Insight 2018;3(14):99327. 10.1172/jci.insight.99327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Staaf J, Jonsson G, Jonsson M, et al. Relation between smoking history and gene expression profiles in lung adenocarcinomas. BMC Med Genomics 2012;5:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Su D, Wang X, Campbell MR, et al. Distinct epigenetic effects of tobacco smoking in whole blood and among leukocyte subtypes. PLoS One 2016;11:e0166486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Bauer M, Fink B, Thurmann L, Eszlinger M, Herberth G, Lehmann I. Tobacco smoking differently influences cell types of the innate and adaptive immune system‐indications from CpG site methylation. Clin Epigenetics 2015;7:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Bauer M, Linsel G, Fink B, et al. A varying T cell subtype explains apparent tobacco smoking induced single CpG hypomethylation in whole blood. Clin Epigenetics 2015;7:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Desrichard A, Kuo F, Chowell D, et al. Tobacco smoking‐associated alterations in the immune microenvironment of squamous cell carcinomas. J Natl Cancer Inst 2018;110:1386–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD‐1 blockade in non‐small cell lung cancer. Science 2015;348:124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Laryngoscope Investigative Otolaryngology are provided here courtesy of Wiley

RESOURCES