[Skip to Navigation]
Sign In
Figure 1.  Box Plot and Unpaired t Test of Tumor Mutational Burden (TMB) by Tumor Grade
Box Plot and Unpaired t Test of Tumor Mutational Burden (TMB) by Tumor Grade

The dark line in each box represents the median TMB; ends of the boxes, 25th and 75th percentile TMB; and whiskers, minimum and maximum TMB for each group. Mut indicates mutation; Mb, megabase.

Figure 2.  Kaplan-Meier Survival Curves by Tumor Grade
Kaplan-Meier Survival Curves by Tumor Grade

HGT indicates high-grade tumors; HR, hazard ratio; LGT, low-grade tumors; OS, overall survival; PFS, progression-free survival.

Table 1.  Baseline Characteristics of All Patients With Mucosal HNSCC According to Tumor Grade
Baseline Characteristics of All Patients With Mucosal HNSCC According to Tumor Grade
Table 2.  Bivariate Analysis for the Association Between Tumor Grade and Complete Response or Partial Response to Immunotherapy
Bivariate Analysis for the Association Between Tumor Grade and Complete Response or Partial Response to Immunotherapy
Table 3.  Univariate and Multivariable Standard Logistic Regression Models for a Clinically Beneficial Immunotherapy Response
Univariate and Multivariable Standard Logistic Regression Models for a Clinically Beneficial Immunotherapy Response
1.
Ferris  RL.  Immunology and immunotherapy of head and neck cancer.   J Clin Oncol. 2015;33(29):3293-3304. doi:10.1200/JCO.2015.61.1509PubMedGoogle ScholarCrossref
2.
Manca  P, Raez  LE, Salzberg  M, Sanchez  J, Hunis  B, Rolfo  C.  The value of immunotherapy in head and neck cancer.   Expert Opin Biol Ther. 2019;19(1):35-43. doi:10.1080/14712598.2019.1556637PubMedGoogle ScholarCrossref
3.
Ferris  RL, Blumenschein  G, 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:10.1016/j.oraloncology.2018.04.008Google ScholarCrossref
4.
Porosnicu  M, Waltonen  JD, Triozzi  P.  Anti-tumor effect of PD-L1 inhibitor durvalumab administered before surgery in patients with HPV+ vs HPV- HNSCC.   Int J Radiat Oncol. 2018;100(5):1316. doi:10.1016/j.ijrobp.2017.12.299Google ScholarCrossref
5.
Schoenfeld  JD, Hanna  GJ, Jo  VY,  et al.  Neoadjuvant nivolumab or nivolumab plus ipilimumab in untreated oral cavity squamous cell carcinoma: a phase 2 open-label randomized clinical trial.   JAMA Oncol. 2020;6(10):1563-1570. doi:10.1001/jamaoncol.2020.2955PubMedGoogle ScholarCrossref
6.
Ferrarotto  R, Bell  D, Rubin  ML,  et al.  Impact of neoadjuvant durvalumab with or without tremelimumab on CD8+ tumor lymphocyte density, safety, and efficacy in patients with oropharynx cancer: CIAO Trial results.   Clin Cancer Res. 2020;26(13):3211-3219. doi:10.1158/1078-0432.CCR-19-3977PubMedGoogle ScholarCrossref
7.
Siu  LL, Even  C, Mesía  R,  et al.  Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: the phase 2 CONDOR randomized clinical trial.   JAMA Oncol. 2019;5(2):195-203. doi:10.1001/jamaoncol.2018.4628PubMedGoogle ScholarCrossref
8.
Ferris  RL, Gonçalves  A, Baxi  SS,  et al.  An open-label, multicohort, phase 1/2 study in patients with virus-associated cancers (CheckMate 358): safety and efficacy of neoadjuvant nivolumab in squamous cell carcinoma of the head and neck (SCCHN).   Ann Oncol. 2017;28(September):v628-v629. doi:10.1093/annonc/mdx440.041Google ScholarCrossref
9.
Mehra  R, Seiwert  TY, Gupta  S,  et al.  Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012.   Br J Cancer. 2018;119(2):153-159. doi:10.1038/s41416-018-0131-9PubMedGoogle ScholarCrossref
10.
Chow  LQM, Haddad  R, Gupta  S,  et al.  Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort.   J Clin Oncol. 2016;34(32):3838-3845. doi:10.1200/JCO.2016.68.1478PubMedGoogle ScholarCrossref
11.
Cohen  EEW, Soulières  D, Le Tourneau  C,  et al; KEYNOTE-040 investigators.  Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study.   Lancet. 2019;393(10167):156-167. doi:10.1016/S0140-6736(18)31999-8PubMedGoogle ScholarCrossref
12.
Bauml  J, Seiwert  TY, Pfister  DG,  et al.  Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study.   J Clin Oncol. 2017;35(14):1542-1549. doi:10.1200/JCO.2016.70.1524PubMedGoogle ScholarCrossref
13.
Uppaluri  R, Campbell  KM, Egloff  AM,  et al.  Neoadjuvant and adjuvant pembrolizumab in resectable locally advanced, human papillomavirus-unrelated head and neck cancer: a multicenter, phase II trial.   Clin Cancer Res. 2020;26(19):5140-5152. doi:10.1158/1078-0432.CCR-20-1695PubMedGoogle ScholarCrossref
14.
Zandberg  DP, Algazi  AP, Jimeno  A,  et al.  Durvalumab for recurrent or metastatic head and neck squamous cell carcinoma: results from a single-arm, phase II study in patients with ≥25% tumour cell PD-L1 expression who have progressed on platinum-based chemotherapy.   Eur J Cancer. 2019;107:142-152. doi:10.1016/j.ejca.2018.11.015PubMedGoogle ScholarCrossref
15.
Colevas  AD, Bahleda  R, Braiteh  F,  et al.  Safety and clinical activity of atezolizumab in head and neck cancer: results from a phase I trial.   Ann Oncol. 2018;29(11):2247-2253. doi:10.1093/annonc/mdy411PubMedGoogle ScholarCrossref
16.
Yarchoan  M, Hopkins  A, Jaffee  EM.  Tumor mutational burden and response rate to PD-1 inhibition.   N Engl J Med. 2017;377(25):2500-2501. doi:10.1056/NEJMc1713444PubMedGoogle ScholarCrossref
17.
Samstein  RM, Lee  C-H, Shoushtari  AN,  et al.  Tumor mutational load predicts survival after immunotherapy across multiple cancer types.   Nat Genet. 2019;51(2):202-206. doi:10.1038/s41588-018-0312-8PubMedGoogle ScholarCrossref
18.
Goodman  AM, Kato  S, Bazhenova  L,  et al.  Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers.   Mol Cancer Ther. 2017;16(11):2598-2608. doi:10.1158/1535-7163.MCT-17-0386PubMedGoogle ScholarCrossref
19.
Verdegaal  EME, de Miranda  NFCC, Visser  M,  et al.  Neoantigen landscape dynamics during human melanoma-T cell interactions.   Nature. 2016;536(7614):91-95. doi:10.1038/nature18945PubMedGoogle ScholarCrossref
20.
Segal  NH, Parsons  DW, Peggs  KS,  et al.  Epitope landscape in breast and colorectal cancer.   Cancer Res. 2008;68(3):889-892. doi:10.1158/0008-5472.CAN-07-3095PubMedGoogle ScholarCrossref
21.
Mcgranahan  N, Furness  AJS, Rosenthal  R,  et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.   Science. 2016;351(6280):1463-1469. doi:10.1126/science.aaf1490Google ScholarCrossref
22.
Schumacher  TN, Schreiber  RD.  Neoantigens in cancer immunotherapy.   Science. 2015;348(6230):69-74. doi:10.1126/science.aaa4971Google ScholarCrossref
23.
Elston  CW, Ellis  IO.  Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: Experience from a large study with long-term follow-up. C. W. Elston & I. O. Ellis. Histopathology 1991; 19; 403-410—author commentary.   Histopathology. 2002;41(3A):151-153. doi:10.1046/j.1365-2559.2002.14691.xGoogle Scholar
24.
Lughezzani  G, Burger  M, Margulis  V,  et al.  Prognostic factors in upper urinary tract urothelial carcinomas: a comprehensive review of the current literature.   Eur Urol. 2012;62(1):100-114. doi:10.1016/j.eururo.2012.02.030PubMedGoogle ScholarCrossref
25.
Mendenhall  WM, Indelicato  DJ, Scarborough  MT,  et al.  The management of adult soft tissue sarcomas.   Am J Clin Oncol. 2009;32(4):436-442. doi:10.1097/COC.0b013e318173a54fPubMedGoogle ScholarCrossref
26.
Kim  JY, Hong  SM, Ro  JY.  Recent updates on grading and classification of neuroendocrine tumors.   Ann Diagn Pathol. 2017;29:11-16. doi:10.1016/j.anndiagpath.2017.04.005PubMedGoogle ScholarCrossref
27.
Weller  M, Wick  W, Aldape  K,  et al.  Glioma.   Nat Rev Dis Primers. 2015;1(July):15017. doi:10.1038/nrdp.2015.17PubMedGoogle ScholarCrossref
28.
Anderson  EM, Luu  M, Balzer  BL,  et al.  Variations in the association of grade with survival across the head and neck cancer landscape.   Head Neck. 2021;43(4):1105-1115. doi:10.1002/hed.26566PubMedGoogle ScholarCrossref
29.
Argiris  A, Li  Y, Forastiere  A.  Prognostic factors and long-term survivorship in patients with recurrent or metastatic carcinoma of the head and neck.   Cancer. 2004;101(10):2222-2229. doi:10.1002/cncr.20640PubMedGoogle ScholarCrossref
30.
Mirabile  A, Miceli  R, Calderone  RG,  et al.  Prognostic factors in recurrent or metastatic squamous cell carcinoma of the head and neck.   Head Neck. 2019;41(6):1895-1902. doi:10.1002/hed.25636PubMedGoogle ScholarCrossref
31.
Thomas  B, Stedman  M, Davies  L.  Grade as a prognostic factor in oral squamous cell carcinoma: a population-based analysis of the data.   Laryngoscope. 2014;124(3):688-694. doi:10.1002/lary.24357PubMedGoogle ScholarCrossref
32.
Mehta  V, Yu  GP, Schantz  SP.  Population-based analysis of oral and oropharyngeal carcinoma: changing trends of histopathologic differentiation, survival and patient demographics.   Laryngoscope. 2010;120(11):2203-2212. doi:10.1002/lary.21129PubMedGoogle ScholarCrossref
33.
Johnson  A, Severson  E, Gay  L,  et al.  Comprehensive genomic profiling of 282 pediatric low- and high-grade gliomas reveals genomic drivers, tumor mutational burden, and hypermutation signatures.   Oncologist. 2017;22(12):1478-1490. doi:10.1634/theoncologist.2017-0242PubMedGoogle ScholarCrossref
34.
Iwabuchi  H, Sakamoto  M, Sakunaga  H,  et al.  Genetic analysis of benign, low-grade, and high-grade ovarian tumors.   Cancer Res. 1995;55(24):6172-6180. PubMedGoogle Scholar
35.
Caso  R, Sanchez-Vega  F, Tan  KS,  et al.  The underlying tumor genomics of predominant histologic subtypes in lung adenocarcinoma.   J Thorac Oncol. 2020;15(12):1844-1856. doi:10.1016/j.jtho.2020.08.005PubMedGoogle ScholarCrossref
36.
Bi  WL, Prabhu  VC, Dunn  IF.  High-grade meningiomas: biology and implications.   Neurosurg Focus. 2018;44(4):E2. doi:10.3171/2017.12.FOCUS17756PubMedGoogle ScholarCrossref
37.
Eisenhauer  EA, Therasse  P, Bogaerts  J,  et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).   Eur J Cancer. 2009;45(2):228-247. doi:10.1016/j.ejca.2008.10.026PubMedGoogle ScholarCrossref
38.
Heinze  G, Schemper  M.  A solution to the problem of separation in logistic regression.   Stat Med. 2002;21(16):2409-2419. doi:10.1002/sim.1047PubMedGoogle ScholarCrossref
Original Investigation
April 28, 2022

Tumor Histological Grade and Immunotherapy Response in Patients With Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma

Author Affiliations
  • 1Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
  • 2Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medical Institutions, Baltimore, Maryland
  • 3Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
  • 4Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
JAMA Otolaryngol Head Neck Surg. 2022;148(6):540-546. doi:10.1001/jamaoto.2022.0640
Key Points

Question  Is tumor histological grade associated with immunotherapy response in patients with recurrent or metastatic mucosal head and neck squamous cell carcinoma?

Findings  In this cohort study of 60 patients with recurrent or metastatic head and neck squamous cell carcinoma treated with immune checkpoint inhibitors, patients with high-grade tumors were more likely to have a clinically beneficial response to immunotherapy than patients with low-grade tumors.

Meaning  These findings suggest that high-grade tumors in recurrent or metastatic head and neck squamous cell carcinoma may have improved immunotherapy response compared with low-grade tumors.

Abstract

Importance  Tumor histological factors that predict immunotherapy response in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) are not well defined.

Objective  To investigate the association between tumor grade and immunotherapy response in patients with recurrent or metastatic mucosal HNSCC.

Design, Setting, and Participants  In this retrospective cohort study, the medical records of 60 patients with recurrent or metastatic mucosal HNSCC treated with immune checkpoint inhibitors at Johns Hopkins Hospital between July 1, 2015, and January 22, 2020, were reviewed.

Exposures  High-grade tumors (HGTs) vs low-grade tumors (LGTs) in recurrent or metastatic HNSCC.

Main Outcomes and Measures  Patients were divided into 2 groups: those with LGTs (well differentiated and moderately differentiated) and those with HGTs (poorly differentiated). The main outcome was a clinically beneficial immunotherapy response, defined as complete response or partial response. Univariable and multivariable logistic regressions were conducted to calculate odds ratios for each variable’s association with immunotherapy response. Survival differences were evaluated using Kaplan-Meier survival curves with multivariable Cox proportional hazards regression models.

Results  The 60 patients (35 with HGTs and 25 with LGTs) had a mean (SD) age of 64.6 (8.88) years; 51 were male (85%); and 38 were current or former smokers (63%). The oropharynx was the most common primary tumor site both in patients with HGTs (22 of 35; 63%) and those with LGTs (12 of 25; 48%). Bivariate analysis showed the proportion of patients having a beneficial response to immunotherapy was greater for patients with HGTs (12 of 35; 34.3%) than those with LGTs (2 of 25, 8.0%) (difference, 26.3%; 95% CI, 7.3%-45.3%). Upon multivariable analysis, patients with HGTs had 5.35-fold increased odds (95% CI, 1.04-27.37) of having a clinically beneficial response to immunotherapy. Among patients with available tumor genomic profiling data, the mean tumor mutational burden was greater for patients with HGTs (mean [SD], 8.6 [5.4] mut/Mb; n = 8) than patients with LGTs (mean [SD], 3.6 [1.1] mut/Mb; n = 4) (difference = 5.0 mut/Mb; 95% CI −1.4 to 11.4 mut/Mb; Cohen d = 1.2).

Conclusions and Relevance  In this cohort study, tumor grade was independently associated with immunotherapy response in patients with recurrent or metastatic mucosal HNSCC. These findings highlight the potential role of tumor grade in predicting immunotherapy response in mucosal HNSCC.

Introduction

The use of immune checkpoint inhibitors (ICIs) has shown promise as an emerging treatment option for patients with advanced head and neck cancer. These ICIs include antibodies that antagonize the PD-1/PD-L1 axis or CTLA-4.1 Previous studies of head and neck squamous cell carcinoma (HNSCC) tumors have demonstrated a clinically beneficial response to ICIs in patients with disease that is refractory to traditional therapies, including surgery, chemotherapy, and radiation therapy.2-15

One biomarker that has been associated with a beneficial response to ICI across solid tumors is tumor mutational burden (TMB).16 This association between increased TMB and a beneficial response to ICI has been demonstrated in numerous cancer types and tumor histologies.17,18 One hypothesis explaining such an association is the higher probability of generating mutation-associated tumor neoantigens with higher TMB. This probability increases the likelihood of immune detection of these foreign tumor neoantigens, thus facilitating a more robust immune response against the tumor cells.19-22

While tumor grade is considered a factor of prognostic and therapeutic significance in numerous cancer types,23-27 its role in mucosal HNSCC is more controversial, and it is therefore not used as a staging criterion.28-32 High-grade tumors (HGTs) proliferate rapidly and display greater degrees of genomic instability, facilitating the accumulation of more mutations and subsequently a higher TMB. Therefore, we hypothesized that tumor histological grade may be associated with responses to an immune checkpoint blockade. Tumor mutational burden has been positively correlated with a beneficial response to ICIs, which implies that HGT may be also associated with a clinically beneficial response to ICIs, thus underscoring its clinical utility as a predictive biomarker. Though similar associations have been demonstrated in other histologies, this association has yet to be characterized in head and neck cancer.33-36 To our knowledge, the association between tumor histological grade and ICI response has not been examined in patients with mucosal HNSCC. The objective of this study is to examine the association between tumor grade and immunotherapy response in patients treated with ICI for recurrent or metastatic mucosal HNSCC.

Methods
Study Participants

This single-institution retrospective cohort study was approved by the Johns Hopkins Hospital institutional review board. Patients treated with ICIs for recurrent or metastatic HNSCC at the Johns Hopkins Hospital between July 1, 2015, and January 22, 2020, were included in the analysis. The ICIs included in this study were pembrolizumab, nivolumab, ipilimumab, and durvalumab. Patients were excluded if they met any of the following exclusion criteria: the ICI was not for a primary mucosal HNSCC tumor; there was an absence of tumor grade data in the patient medical records; the patient died prior to imaging; the patient was under 18 years of age; the patient was lost to follow-up; or the patient had an unknown primary tumor. This study followed the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) reporting guideline for cohort studies.

Clinical Data

The electronic medical records of eligible patients were reviewed. Baseline patient characteristics at immunotherapy start date included age, sex, race and ethnicity, body mass index (BMI; calculated as weight in kilograms divided by height in meters squared), smoking history, and human papillomavirus (HPV) tumor status. Patients’ HPV status was determined via p16 immunohistochemical analysis or in situ hybridization. Cancer characteristics included the primary tumor site, tumor grade, T-stage, and the sites and total number of distant organ metastases. Well-differentiated and moderately differentiated tumors were classified as low-grade tumors (LGTs), and poorly differentiated tumors were classified as HGTs; these served as the 2 comparison groups in this study. There were no patients in this cohort with undifferentiated tumors. Clinician progress notes and radiology reports were reviewed to collect data on disease progression or remission after treatment. Patients whose cancers had a complete response or partial response according to the RECIST guidelines (version 1.1) were defined as having a clinical benefit from immunotherapy.37 Patients with progressive disease or stable disease after treatment with an ICI were considered immunotherapy nonresponders.

Statistical Analysis

All statistical analyses were completed using Stata, version 15.1 (StataCorp LLC). Univariate and multivariable standard logistic regressions were conducted to calculate odds ratios (ORs) for the association of each variable with a clinically beneficial immunotherapy response. Tumor grade and HPV status were adjusted for in the multivariable model. Owing to small sample size leading to a separation of data for patients with hypopharyngeal tumors (n = 4) in the standard logistic regression model, these patients were excluded from the standard logistic regression, and a Firth’s logistic regression including these patients was subsequently conducted to serve as a sensitivity analysis. Kaplan-Meier analyses were performed to assess progression-free survival (PFS) and overall survival (OS), and median survival times with 95% CIs were calculated for the LGT and HGT groups. Multivariable Cox proportional hazards regression models for PFS and OS was used to generate adjusted hazard ratios (HRs); consistent with the standard logistic regression, the 4 patients with hypopharyngeal tumors were excluded from the multivariable Cox proportional hazards regression models. Results were reported with appropriate effect size, and 95% CIs were used to define the precision of the estimates.

Results
Baseline Characteristics

The baseline characteristics of the study cohort are summarized in Table 1. Sixty patients were analyzed, including 25 with LGTs and 35 with HGTs. The mean (SD) age was 64.6 (8.88) years. Fifty-one patients were male (85%), and 9 were female (15%); 1 was Asian (2%), 12 were Black (20%), 1 was Hispanic (2%), and 46 were White (77%). Thirty-eight patients (63%) had a current or former smoking status. The most common primary tumor site was the oropharynx in patients with LGTs (12 of 25; 48%) and in those with HGTs (22 of 35; 63%). The HPV positivity rate was 32% (8 of 25) in patients with LGTs and 54% (19 of 35) in patients with HGT (difference of 22%). Bivariate analyses showed that the HGT and LGT groups were similar in mean age, sex, race, BMI, smoking status, primary tumor site, HPV status, presenting T-stage, presenting N-stage, and metastases to the lung, bone, and liver. Prior to treatment with ICIs, patients most commonly received surgery, chemotherapy, and radiation therapy (26 of 60; 43%) or chemotherapy and radiation therapy (28 of 60; 47%) (eTable 1 in the Supplement).

Bivariate Analyses of Tumor Grade and Immunotherapy Response

The clinically beneficial immunotherapy response rate, defined as complete response or partial response, was 23.3% (14 of 60) (Table 2). Bivariate analysis showed that patients with HGTs were more likely to have a clinically beneficial immunotherapy response (12 of 35; 34.3%) than those with LGTs (2 of 25; 8.0%) (difference, 26.3%; 95% CI, 7.3%-45.3%). When stratifying by tumor grade, the proportion of patients having a clinically beneficial immunotherapy response increased with increasing grade: 0% of well-differentiated tumors, 10.5% of moderately differentiated tumors, and 34.3% of poorly differentiated tumors had a beneficial response (eTable 2 in the Supplement). Progressive disease occurred at a higher rate in patients with LGTs (17 of 25; 68%) than HGTs (10 of 35; 28.6%) (eTable 3 in the Supplement).

Univariate and Multivariable Analyses

Univariate analysis found that patients with HGT had 6.0 times increased odds of having a clinically beneficial immunotherapy response (95% CI, 1.19-30.15) compared with patients with LGTs (Table 3). Multivariable analysis was performed, adjusting for tumor grade and HPV status. Upon multivariable analysis, HGTs (adjusted OR, 5.35; 95% CI, 1.04-27.37) remained associated with increased odds of a clinically beneficial immunotherapy response. Age, sex, primary tumor site, HPV status, smoking history, T-stage, total number of metastases, and presence of metastases to the lung, bone, or liver were not associated with increased or decreased odds of a clinically beneficial immunotherapy response in univariable or multivariable analyses.

Tumor Mutational Burden Subgroup Analysis

Within the study cohort, there were 4 patients with LGTs and 8 patients with HGTs with available TMB data. Figure 1 depicts the distribution of TMB for patients with LGTs and HGTs. Among patients with available TMB data, the mean TMB was greater for patients with HGTs (mean [SD], 8.6 [5.4] mut/Mb; n = 8) than LGTs (mean [SD], 3.6 [1.1] mut/Mb; n = 4) (difference, 5.0 mut/Mb; 95% CI, −1.4 to 11.4; Cohen d = 1.2). The HGT group also had a higher median TMB than the LGT group (median = 10.4 mut/Mb [IQR, 3.0-12.0 mut/Mb] and median = 3.5 mut/Mb [IQR, 2.5-5.0 mut/Mb], respectively).

Progression-Free Survival and Overall Survival
Full Cohort by Tumor Grade

Across the full study cohort, the median PFS was 5.9 months (95% CI, 3.8-11.1 months) for patients with HGTs (n = 33) vs 3.3 months (95% CI, 1.7-4.7 months) for patients with LGTs (n = 23). Multivariable analysis showed a decreased risk of disease progression for patients with HGTs compared with LGTs (adjusted HR, 0.69; 95% CI, 0.39-1.22) (Figure 2A). The median OS for patients with HGTs vs LGTs was 16.6 months (95% CI, 11.1-25.0 months) vs 15.0 months (95% CI, 8.2-22.8 months). The risk of death was slightly lower in the HGT group compared with the LGT group (adjusted HR, 0.94; 95% CI, 0.47-1.89) (Figure 2B).

Discussion

Tumor grade is associated with poorer prognosis in numerous cancer types; however, its prognostic value is regarded as less pertinent in HNSCC. This cohort study demonstrates an increased response to ICIs in patients with HGTs compared with LGTs in mucosal HNSCC. To our knowledge, these findings are the first to demonstrate this association in HNSCC. Unlike genomic biomarkers for response to immunotherapy, such as TMB, which can be costly and not easily available, tumor grade is routinely evaluated for head and neck tumors and is readily available to physicians. This availability underscores the clinical utility of tumor grade as potential predictive biomarker in the clinical setting to help guide treatment decisions and patient counseling.

On multivariable analysis, patients with HGTs had increased odds of having a clinically beneficial immunotherapy response. The magnitude of this OR was greater than any of the other covariates assessed (primary tumor site, T-stage, site and number of distant organ metastases, sex, age, BMI, HPV-status, and smoking history). A sensitivity analysis adjusting for the presence of advanced nodal disease was performed and did not alter the results or findings presented in Table 3. Additional sensitivity analyses adjusting for tumor grade, primary tumor site, HPV status, smoking status, and BMI were also performed and did not meaningfully alter the results or findings presented in Table 3. Likely limited by small sample bias, none of the 4 patients with primary tumors of the hypopharynx had a clinically beneficial response to immunotherapy. Thus, these patients were excluded from the standard logistic regression, as it is not possible to produce an OR estimate when there is complete or quasicomplete separation of the data.38 A Firth’s logistic regression that included these 4 patients was conducted to serve as a sensitivity analysis for the standard logistic regression (eTable 4 in the Supplement). Firth’s logistic regression supported the same findings as those shown in Table 3, and the magnitudes of ORs for all variables were comparable. It is notable that numerous ORs in Table 3 have wide confidence intervals owing to small sample size. Though Table 3 suggests that HGTs are associated with increased odds of a clinically beneficial immunotherapy response, a definitive conclusion cannot be drawn from these data.

Tumor Mutational Burden

With increased TMB having been shown to be associated with improved ICI response,16-18 it is notable that patients with HGTs had a higher mean TMB compared with patients with LGTs. Owing to the limited subset of patients for whom TMB data were available, analyses of tumor grade with immunotherapy response while controlling for TMB in both groups could not be reliably conducted. Furthermore, as increasing tumor grade contributes to genomic instability and resultant increased tumor mutational burden, tumor grade and TMB are not entirely independent variables.

Tumor mutational burden is commonly used as a biomarker to estimate response to treatment with ICIs. If a potential association between TMB and tumor grade could be demonstrated in a larger cohort of patients, it is possible that tumor grade may be used as an inexpensive and readily available, albeit imperfect, surrogate method for estimating low vs high TMB. Although no studies have examined the association between TMB and tumor grade in patients with mucosal HNSCC primary tumors, Johnson et al33 demonstrated in a comparison of 157 patients with high-grade gliomas and 125 patients with low-grade gliomas that high-grade gliomas had a greater median TMB (1.8 mut/Mb vs 0.9 mut/Mb for high-grade vs low-grade gliomas, respectively) and percentage of patients with intermediate TMB (6-19 mut/Mb; 12.1% vs 3.2% for high-grade vs low-grade gliomas, respectively) and high TMB (≥20 mut/Mb; 5.7% vs 0.8% for high-grade vs low-grade gliomas, respectively). Similarly, this association between high-grade tumors and increased tumor mutations has been shown in numerous other tumor types, such as ovarian tumors,34 lung adenocarcinoma,35 and meningiomas.36

Survival Analyses

Because of the small sample size of immunotherapy responders included in the Kaplan-Meier survival analyses, we are limited in the conclusions and comparisons we can make between groups regarding PFS and OS. Although patients with HGTs had a markedly higher response rate to ICIs compared with those with LGTs, Kaplan-Meier survival analyses suggested that patients with HGTs had slightly increased median PFS and OS compared with those with HGTs. Cox regression analysis suggests that HGTs may confer a protective effect against disease progression in patients with immunotherapy; however, the confidence interval spanning across 1.0 and the small sample size limit any definitive conclusion being made from these data. Although our study is sufficiently powered to detect a robust difference in clinical response rate to immunotherapy between HGTs and LGTs, it may not be sufficiently powered to detect potential survival differences. As data mature and additional cohorts of patients with greater sample sizes are analyzed, meaningful differences in survival may be observable.

Limitations

Although the findings and implications of this study are promising, it is important to acknowledge and discuss the study’s limitations as well. First, this study is limited by its small sample size and small number of patients who experienced a clinically beneficial immunotherapy response, which limits the number of independent variables that can be incorporated in the multivariable models as well as our ability to draw any meaningful conclusions for PFS and OS differences between the HGT and LGT groups. Additionally, this study is an observational study and thus can establish correlation but not direct causality. The lack of control group further limits the interpretation of the results. Furthermore, the retrospective design of the study depends on the accuracy of documentation in the patients’ electronic medical records. In addition, this is a single-institution study conducted at a tertiary care center, which may limit the generalizability of the findings. The presence of TMB data in only a subgroup of patients limits our ability to draw any conclusions regarding any potential association between tumor grade and TMB. Finally, these patients had differing sites and patterns of metastasis, further contributing to intragroup and intergroup heterogeneity.

Conclusions

In this cohort study, we found that tumor grade was independently associated with increased odds of having an immunotherapy response in patients with recurrent or metastatic primary mucosal HNSCC. As immunotherapy continues to be used in the treatment of patients with recurrent or metastatic HNSCC, these findings suggest the potential role of tumor grade in estimating immunotherapy response. This study also highlights the need for additional confirmatory studies to further define the association between tumor grade, tumor mutational burden, and response to immunotherapy in head and neck cancer.

Back to top
Article Information

Accepted for Publication: March 12, 2022.

Published Online: April 28, 2022. doi:10.1001/jamaoto.2022.0640

Corresponding Author: Rajarsi Mandal, MD, Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, Sixth Floor, Baltimore, MD 21287 (rmandal6@jhmi.edu).

Author Contributions: Mr Alkhatib and Dr Mandal had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Concept and design: Alkhatib, Zhu, Guller, Pardoll, Mandal.

Acquisition, analysis, or interpretation of data: Alkhatib, Maroun, Amin, Zhu, Guller, Herberg, Wu, Seiwert, Rooper, Eisele, Fakhry, Mandal.

Drafting of the manuscript: Alkhatib, Amin, Guller, Mandal.

Critical revision of the manuscript for important intellectual content: All authors.

Statistical analysis: Alkhatib, Zhu, Guller.

Obtained funding: Mandal.

Administrative, technical, or material support: Maroun, Zhu, Guller, Herberg, Wu, Seiwert, Fakhry, Mandal.

Supervision: Maroun, Eisele, Pardoll, Mandal.

Other: Rooper.

Conflict of Interest Disclosures: Dr Wu reported receiving personal fees from AstraZeneca outside the submitted work. Dr Seiwert reported receiving grants and institutional trial funding from Merck, Bristol Myers Squibb, CUE Biopharma, AstraZeneca, Roche, Nanobiotix and advisory board honoraria from Merck, Vir Biotechnology, Innate, Nanobiotix, and Kura outside the submitted work. No other disclosures were reported.

References
1.
Ferris  RL.  Immunology and immunotherapy of head and neck cancer.   J Clin Oncol. 2015;33(29):3293-3304. doi:10.1200/JCO.2015.61.1509PubMedGoogle ScholarCrossref
2.
Manca  P, Raez  LE, Salzberg  M, Sanchez  J, Hunis  B, Rolfo  C.  The value of immunotherapy in head and neck cancer.   Expert Opin Biol Ther. 2019;19(1):35-43. doi:10.1080/14712598.2019.1556637PubMedGoogle ScholarCrossref
3.
Ferris  RL, Blumenschein  G, 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:10.1016/j.oraloncology.2018.04.008Google ScholarCrossref
4.
Porosnicu  M, Waltonen  JD, Triozzi  P.  Anti-tumor effect of PD-L1 inhibitor durvalumab administered before surgery in patients with HPV+ vs HPV- HNSCC.   Int J Radiat Oncol. 2018;100(5):1316. doi:10.1016/j.ijrobp.2017.12.299Google ScholarCrossref
5.
Schoenfeld  JD, Hanna  GJ, Jo  VY,  et al.  Neoadjuvant nivolumab or nivolumab plus ipilimumab in untreated oral cavity squamous cell carcinoma: a phase 2 open-label randomized clinical trial.   JAMA Oncol. 2020;6(10):1563-1570. doi:10.1001/jamaoncol.2020.2955PubMedGoogle ScholarCrossref
6.
Ferrarotto  R, Bell  D, Rubin  ML,  et al.  Impact of neoadjuvant durvalumab with or without tremelimumab on CD8+ tumor lymphocyte density, safety, and efficacy in patients with oropharynx cancer: CIAO Trial results.   Clin Cancer Res. 2020;26(13):3211-3219. doi:10.1158/1078-0432.CCR-19-3977PubMedGoogle ScholarCrossref
7.
Siu  LL, Even  C, Mesía  R,  et al.  Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: the phase 2 CONDOR randomized clinical trial.   JAMA Oncol. 2019;5(2):195-203. doi:10.1001/jamaoncol.2018.4628PubMedGoogle ScholarCrossref
8.
Ferris  RL, Gonçalves  A, Baxi  SS,  et al.  An open-label, multicohort, phase 1/2 study in patients with virus-associated cancers (CheckMate 358): safety and efficacy of neoadjuvant nivolumab in squamous cell carcinoma of the head and neck (SCCHN).   Ann Oncol. 2017;28(September):v628-v629. doi:10.1093/annonc/mdx440.041Google ScholarCrossref
9.
Mehra  R, Seiwert  TY, Gupta  S,  et al.  Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012.   Br J Cancer. 2018;119(2):153-159. doi:10.1038/s41416-018-0131-9PubMedGoogle ScholarCrossref
10.
Chow  LQM, Haddad  R, Gupta  S,  et al.  Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort.   J Clin Oncol. 2016;34(32):3838-3845. doi:10.1200/JCO.2016.68.1478PubMedGoogle ScholarCrossref
11.
Cohen  EEW, Soulières  D, Le Tourneau  C,  et al; KEYNOTE-040 investigators.  Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study.   Lancet. 2019;393(10167):156-167. doi:10.1016/S0140-6736(18)31999-8PubMedGoogle ScholarCrossref
12.
Bauml  J, Seiwert  TY, Pfister  DG,  et al.  Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study.   J Clin Oncol. 2017;35(14):1542-1549. doi:10.1200/JCO.2016.70.1524PubMedGoogle ScholarCrossref
13.
Uppaluri  R, Campbell  KM, Egloff  AM,  et al.  Neoadjuvant and adjuvant pembrolizumab in resectable locally advanced, human papillomavirus-unrelated head and neck cancer: a multicenter, phase II trial.   Clin Cancer Res. 2020;26(19):5140-5152. doi:10.1158/1078-0432.CCR-20-1695PubMedGoogle ScholarCrossref
14.
Zandberg  DP, Algazi  AP, Jimeno  A,  et al.  Durvalumab for recurrent or metastatic head and neck squamous cell carcinoma: results from a single-arm, phase II study in patients with ≥25% tumour cell PD-L1 expression who have progressed on platinum-based chemotherapy.   Eur J Cancer. 2019;107:142-152. doi:10.1016/j.ejca.2018.11.015PubMedGoogle ScholarCrossref
15.
Colevas  AD, Bahleda  R, Braiteh  F,  et al.  Safety and clinical activity of atezolizumab in head and neck cancer: results from a phase I trial.   Ann Oncol. 2018;29(11):2247-2253. doi:10.1093/annonc/mdy411PubMedGoogle ScholarCrossref
16.
Yarchoan  M, Hopkins  A, Jaffee  EM.  Tumor mutational burden and response rate to PD-1 inhibition.   N Engl J Med. 2017;377(25):2500-2501. doi:10.1056/NEJMc1713444PubMedGoogle ScholarCrossref
17.
Samstein  RM, Lee  C-H, Shoushtari  AN,  et al.  Tumor mutational load predicts survival after immunotherapy across multiple cancer types.   Nat Genet. 2019;51(2):202-206. doi:10.1038/s41588-018-0312-8PubMedGoogle ScholarCrossref
18.
Goodman  AM, Kato  S, Bazhenova  L,  et al.  Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers.   Mol Cancer Ther. 2017;16(11):2598-2608. doi:10.1158/1535-7163.MCT-17-0386PubMedGoogle ScholarCrossref
19.
Verdegaal  EME, de Miranda  NFCC, Visser  M,  et al.  Neoantigen landscape dynamics during human melanoma-T cell interactions.   Nature. 2016;536(7614):91-95. doi:10.1038/nature18945PubMedGoogle ScholarCrossref
20.
Segal  NH, Parsons  DW, Peggs  KS,  et al.  Epitope landscape in breast and colorectal cancer.   Cancer Res. 2008;68(3):889-892. doi:10.1158/0008-5472.CAN-07-3095PubMedGoogle ScholarCrossref
21.
Mcgranahan  N, Furness  AJS, Rosenthal  R,  et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.   Science. 2016;351(6280):1463-1469. doi:10.1126/science.aaf1490Google ScholarCrossref
22.
Schumacher  TN, Schreiber  RD.  Neoantigens in cancer immunotherapy.   Science. 2015;348(6230):69-74. doi:10.1126/science.aaa4971Google ScholarCrossref
23.
Elston  CW, Ellis  IO.  Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: Experience from a large study with long-term follow-up. C. W. Elston & I. O. Ellis. Histopathology 1991; 19; 403-410—author commentary.   Histopathology. 2002;41(3A):151-153. doi:10.1046/j.1365-2559.2002.14691.xGoogle Scholar
24.
Lughezzani  G, Burger  M, Margulis  V,  et al.  Prognostic factors in upper urinary tract urothelial carcinomas: a comprehensive review of the current literature.   Eur Urol. 2012;62(1):100-114. doi:10.1016/j.eururo.2012.02.030PubMedGoogle ScholarCrossref
25.
Mendenhall  WM, Indelicato  DJ, Scarborough  MT,  et al.  The management of adult soft tissue sarcomas.   Am J Clin Oncol. 2009;32(4):436-442. doi:10.1097/COC.0b013e318173a54fPubMedGoogle ScholarCrossref
26.
Kim  JY, Hong  SM, Ro  JY.  Recent updates on grading and classification of neuroendocrine tumors.   Ann Diagn Pathol. 2017;29:11-16. doi:10.1016/j.anndiagpath.2017.04.005PubMedGoogle ScholarCrossref
27.
Weller  M, Wick  W, Aldape  K,  et al.  Glioma.   Nat Rev Dis Primers. 2015;1(July):15017. doi:10.1038/nrdp.2015.17PubMedGoogle ScholarCrossref
28.
Anderson  EM, Luu  M, Balzer  BL,  et al.  Variations in the association of grade with survival across the head and neck cancer landscape.   Head Neck. 2021;43(4):1105-1115. doi:10.1002/hed.26566PubMedGoogle ScholarCrossref
29.
Argiris  A, Li  Y, Forastiere  A.  Prognostic factors and long-term survivorship in patients with recurrent or metastatic carcinoma of the head and neck.   Cancer. 2004;101(10):2222-2229. doi:10.1002/cncr.20640PubMedGoogle ScholarCrossref
30.
Mirabile  A, Miceli  R, Calderone  RG,  et al.  Prognostic factors in recurrent or metastatic squamous cell carcinoma of the head and neck.   Head Neck. 2019;41(6):1895-1902. doi:10.1002/hed.25636PubMedGoogle ScholarCrossref
31.
Thomas  B, Stedman  M, Davies  L.  Grade as a prognostic factor in oral squamous cell carcinoma: a population-based analysis of the data.   Laryngoscope. 2014;124(3):688-694. doi:10.1002/lary.24357PubMedGoogle ScholarCrossref
32.
Mehta  V, Yu  GP, Schantz  SP.  Population-based analysis of oral and oropharyngeal carcinoma: changing trends of histopathologic differentiation, survival and patient demographics.   Laryngoscope. 2010;120(11):2203-2212. doi:10.1002/lary.21129PubMedGoogle ScholarCrossref
33.
Johnson  A, Severson  E, Gay  L,  et al.  Comprehensive genomic profiling of 282 pediatric low- and high-grade gliomas reveals genomic drivers, tumor mutational burden, and hypermutation signatures.   Oncologist. 2017;22(12):1478-1490. doi:10.1634/theoncologist.2017-0242PubMedGoogle ScholarCrossref
34.
Iwabuchi  H, Sakamoto  M, Sakunaga  H,  et al.  Genetic analysis of benign, low-grade, and high-grade ovarian tumors.   Cancer Res. 1995;55(24):6172-6180. PubMedGoogle Scholar
35.
Caso  R, Sanchez-Vega  F, Tan  KS,  et al.  The underlying tumor genomics of predominant histologic subtypes in lung adenocarcinoma.   J Thorac Oncol. 2020;15(12):1844-1856. doi:10.1016/j.jtho.2020.08.005PubMedGoogle ScholarCrossref
36.
Bi  WL, Prabhu  VC, Dunn  IF.  High-grade meningiomas: biology and implications.   Neurosurg Focus. 2018;44(4):E2. doi:10.3171/2017.12.FOCUS17756PubMedGoogle ScholarCrossref
37.
Eisenhauer  EA, Therasse  P, Bogaerts  J,  et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).   Eur J Cancer. 2009;45(2):228-247. doi:10.1016/j.ejca.2008.10.026PubMedGoogle ScholarCrossref
38.
Heinze  G, Schemper  M.  A solution to the problem of separation in logistic regression.   Stat Med. 2002;21(16):2409-2419. doi:10.1002/sim.1047PubMedGoogle ScholarCrossref
×