Next Article in Journal
Cancer Patients’ Experiences with Telehealth before and during the COVID-19 Pandemic in British Columbia
Next Article in Special Issue
The Effect of Tobacco Smoke N-Nitrosamines, NNK and NDEA, and Nicotine, on DNA Mismatch Repair Mechanism and miRNA Markers, in Hypopharyngeal Squamous Cell Carcinoma: An In Vivo Model and Clinical Evidence
Previous Article in Journal
A Case Study of Clinical Response to Rucaparib in a Patient with Metastatic Castration-Resistant Prostate Cancer and a RAD51B Alteration
Previous Article in Special Issue
Micro-RNAs, the Cornerstones of the Future of Radiobiology in Head and Neck Cancers?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma

by
Jong Chul Park
1,
Hari N. Krishnakumar
2 and
Srinivas Vinod Saladi
3,*
1
Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
2
Long School of Medicine, UT Health, San Antonio, TX 78229, USA
3
Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(6), 4185-4198; https://doi.org/10.3390/curroncol29060334
Submission received: 23 April 2022 / Revised: 2 June 2022 / Accepted: 7 June 2022 / Published: 8 June 2022
(This article belongs to the Special Issue Advances in Squamous Cell Carcinoma of the Head and Neck)

Abstract

:
With the introduction of immunotherapy, significant improvement has been made in the treatment of head and neck squamous cell carcinoma (HNSCC). However, only a small subset of patients with HNSCC benefit from immunotherapy. The current biomarker, a programmed cell death protein ligand 1 (PD-L1) expression that is widely used in treatment decision making for advanced HNSCC, has only a moderate predictive value. Additionally, PD-L1-based assay has critical inherent limitations due to its highly dynamic nature and lack of standardization. With the advance in molecular techniques and our understanding of biology, more reliable, reproducible, and practical novel biomarkers are being developed. These include but are not limited to neoantigen/mutation characteristics, immune transcriptomes, tumor-infiltrating immune cell composition, cancer epigenomic, proteomics and metabolic characteristics, and plasma-based and organoid assays.

1. Introduction

With approximately 890,000 new cases and 450,000 deaths annually, head and neck squamous cell carcinoma (HNSCC) is the 6th most common cancer globally [1]. HNSCCs are malignant cancers that develop along the superficial squamous layer of mucosal epithelium found in the oral cavity, pharynx, larynx, paranasal sinuses, and nasal cavity [1]. Risk factors for HNSCC include the use of tobacco products, alcohol consumption, and viral infections such as human papillomavirus (HPV) and Epstein-Barr virus (EBV) [2]. About 90% of HNSCC presented with a local or locoregional disease, but 10% are diagnosed at an advanced stage with metastatic disease [3]. For local or locoregional diseases, curative surgery and/or radiation therapy are the mainstream therapy [4]. Unfortunately, 50% of patients develop recurrences after the curative-intent treatment, and these recurrences are often not amenable to curative intent salvage therapy and require palliative systemic therapy [5].
Over the past decade, there has been a significant advance in systemic therapy for advanced HNSCC with the introduction of immunotherapy [6]. Immunotherapeutic agents that block immune checkpoints, such as programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have shown promising efficacy with durable antitumor control in a variety of tumors. In HNSCC, immune checkpoint inhibitors (ICIs) that target PD-1, pembrolizumab and nivolumab are currently used in metastatic or recurrent (R/M) HNSCC based on survival benefits [7]. However, only a small subset of patients with advanced HNSCC benefit, with an objective response rate (ORR) ranging from 15–20% and a large proportion of patients suffer from immune-related toxicity from ICI therapy without clinical benefit. As such, identifying reliable and practical predictive biomarkers for optimal patient selection and improved treatment strategy is highly warranted. This review will discuss the current biomarkers data tested in HNSCC and emerging novel biomarkers.

2. Immunotherapy in HNSCC

In HNSCC, immune checkpoint blocking agents pembrolizumab and nivolumab are currently used for the treatment of advanced HNSCC. Both drugs are monoclonal antibodies (mAb) that block PD-1 preventing interaction with PD-L1 and PD-L2 proteins that inhibit T lymphocyte proliferation, and effector functions, and induce apoptosis of tumor-specific T cells [8]. In 2016, both pembrolizumab and nivolumab were approved for the treatment of R/M HNSCC after platinum-based chemotherapy based on KEYNOTE-040 and CHECKMATE-141 studies, respectively, which demonstrated the survival benefit of anti-PD-1 therapy compared to standard chemotherapy [9,10]. Subsequently, in 2019, the FDA granted approval for the use of pembrolizumab as first-line treatment for patients with R/M HNSCC alone or in combination with chemotherapy based on the KEYNOTE-048 study [11]. These ICIs demonstrated durable anti-tumor activity in patients who responded to therapy, with 85% of responses lasting at least 6 months and 71% lasting for over a year [12]. However, the ORR in studied populations was less than 20% [13].

3. Immune Biomarkers in HNSCC

3.1. PD-L1 Expression

Although the introduction of PD-1 inhibitors has increased the utility of immunotherapy for HNSCC, there is still a significant need for more specific biomarkers that can improve the predictive value for ICI response [14]. When considering the general efficacy of anti-PD-1/PD-L1 therapies, it is widely seen that these therapies have a greater response rate in tumors expressing PD-L1 (PD-L1+) than tumors without PD-L1 expression (PD-L1−) across various forms of anti-PD-1/PD-L1 inhibitor therapies and various tumor types [15,16]. The degree of PD-L1 expression within the tumor microenvironment is represented by the following scoring systems: tumor proportion score (TPS), tumor-infiltrating immune cells (IC), and combined positive score (CPS). TPS is the percentage of viable tumor cells that show partial or complete membrane staining for PD-L1 at any intensity. IC is representative of immune cells infiltrating the tumor site, such as T cells, antigen-presenting cells (APCs), and natural killer (NK) cells that show partial or complete membrane staining for PD-L1 at any intensity [17]. CPS is a combination of TPS and IC that analyzes both tumor cells and infiltrating immune cells in the tumor microenvironment that show partial or complete staining for PD-L1 at any intensity [18]. To calculate a CPS, the pathologist must score the number of PD-L1-positive cells (tumor cells, lymphocytes, and macrophages), divide that total by the number of viable tumor cells, and multiply by 100.

3.2. PD-L1 Biomarker Data in HNSCC

PD-L1 expression has been studied and correlated with clinical outcomes across various anti-PD-1/PD-L1 clinical trials (Table 1). Phase 1 KEYNOTE-012 trial studied the efficacy and safety of pembrolizumab in multiple advanced solid tumors including HNSCC. Within this study, a biomarker analysis of PD-L1 expression was conducted by an immunohistochemistry assay (IHC) using the 22C3 antibody. Of 188 patients with HNSCC, both TPS and CPS with a cutoff of 1 were examined for correlation with response to pembrolizumab. Positive CPS (81% of patients) was correlated with higher ORR, progression-free survival (PFS) rates, and overall survival (OS), but such correlation was not observed with positive TPS tumors (65% of patients) indicating a superior predictive value of CPS [19,20]. The study showed that pembrolizumab had a durable anti-tumor effect on responders while exhibiting a manageable safety profile (43), which was the basis for the accelerated approval of pembrolizumab by the food and drug administration (FDA). These results were also corroborated by the phase 2 KEYNOTE-055, which also supported the use of pembrolizumab in patients with pretreated R/M HNSCC [19,21]. Using a 22C3 assay with a CPS cutoff of 1%, ORR was found to be marginally higher among patients with PD-L1+ tumors (18%) when compared to patients with PD-L1− tumors (12%). Additionally, patients with PD-L1+ tumors had slightly higher 6-month PFS (24%; CPS ≥ 1% and 31%; CPS ≥ 50%) compared to PD-L1− patients (20%; CPS < 1% and 20%; CPS < 50%) and comparable rates of OS at 6 months (59%; CPS ≥ 1% and 60%; CPS ≥ 50% vs. 56%; CPS < 1% and 58%; CPS < 50%) [22].
Atezolizumab, a mAb against PD-L1, was tested in patients with advanced HNSCC. In a phase I study, 32 R/M HNSCC patients were treated with atezolizumab and 22% of them achieved objective responses [33]. In a subgroup analysis based on the PD-L1 expression in the tumor-infiltrating immune cell (IC), the PD-L1 + group (n = 25) defined by IC2/3 (≥5%) by the SP142 Ventana assay had a higher ORR (24%) than PD-L1 IC0/1 (<5%) cohort (ORR 14%, n = 7).
The randomized phase III KEYNOTE-040 further studied the efficacy of pembrolizumab among patients with R/M HNSCC who had already undergone platinum-based chemotherapy, comparing it to standard systemic therapy with the OS as a primary endpoint [23,24]. Pembrolizumab demonstrated superior survival compared to the control arm in this population (hazard ratio (HR) 0.80, p = 0.016). Although this study substantiated prior trends by showing increased rates of survival in PD-L1+ patients, the PD-L1− based biomarker analysis results of this study were different from those of prior studies; the magnitude of survival benefit of pembrolizumab compared to standard therapy was greater in patients with TPS > 50% (HR 0.53) than patients with CPS > 1 (HR 0.74) [25]. Furthermore, a PFS in patients with CPS > 1 was not different compared to the control group while patients with TPS > 50 had a longer PFS than the control group. Nevertheless, when compared to standard therapies, patients who received pembrolizumab had a greater median OS (mOS) regardless of PD-L1 status and the magnitude of survival benefit was even greater in patients with CPS > 1 and TPS > 50% [11].
The CHECKMATE-141 was a phase III randomized study that examined the efficacy of nivolumab as a post-platinum therapy in patients with R/M HNSCC [26]. This study distinguished PD-L1 positivity by tumor expression (TC) ≥ 1% using the 28–8 pharmDx assay. Nivolumab demonstrated a significant increase in OS compared to the standard therapy (HR 0.70, p = 0.01) irrespective of PD-L1 tumor expression or HPV status, with estimated 24-month OS nearly tripling from 6.0% to 16.9% for patients on nivolumab; however, the survival benefit was greater with nivolumab in the PD-L1+ subgroup, 57.3% of all PD-L1 evaluable patients, (HR 0.55) compared to PD-L1− population (HR 0.89) [26]. Notably, the magnitudes of survival benefits in subgroups of patients with tumor PD-L1 levels of ≥5% or ≥10% were similar to that with tumor PD-L1 level ≥ 1% with HR of 0.50 and 0.57, respectively.
KEYNOTE-048 study evaluated pembrolizumab as first-line therapy in R/M HNSCC, examining and comparing outcomes of pembrolizumab alone and in combination with platinum-based chemotherapy to the conventional platinum-based combination chemotherapy and cetuximab (Extreme regimen) using co-primary endpoints of OS and PFS [34]. This study used CPS by the 22C3 assay as the PD-L1 expression measure using 2 cut-off values of 1% and 20% [27]. The results of the study supported the efficacy and safety of pembrolizumab alone or in combination with chemotherapy as an appropriate first-line treatment for R/M HNSCC [28,29]. Treatment with pembrolizumab was non-inferior but did not meet the threshold for superiority in survival compared to the extreme regimen in the total population (HR 0.83, p = 0.0199). However, pembrolizumab alone demonstrated superior survival in tumors with CPS ≥ 20 (HR 0.61, p = 0.0007) and CPS ≥ 1 (HR 0.78, p = 0.0086). On the other hand, pembrolizumab with chemotherapy improved overall survival compared to the extreme regimen in the overall population (HR 0.77, p = 0.0034). Again, the survival benefits were greater with higher PD-L1 expression; HR 0.60 in CPS ≥ 20 (p = 0.0004) and HR 0.65 in CPS ≥ 1 (p < 0.0001).
Durvalumab is a mAb against PD-L1, which is currently used for the treatment of lung cancers. In a phase I/II study evaluating durvalumab single agents in multiple solid tumors, an ORR of 6.5% was seen in the HNSCC cohort (n = 62). This study defined the PD-L1 positive population as PD-L1 expression in ≥25% tumor cells (TC) by SP263 Ventana assay. Durvalumab achieved superior objective response and survival in patients with PD-L1+ HNSCC (ORR = 15%; mOS = 8.4 months) than in patients with PD-L1− tumors (ORR = 2.6%; mOS = 7.4 months) [35]. The subsequent phase II study (HAWK) evaluated durvalumab monotherapy in patients with PD-L1-high R/M HNSCC defined as TC ≥ 25% and demonstrated an ORR of 16.2% and a 12-month survival rate of 33.6% [30]. On the other hand, the CONDOR study evaluated durvalumab along with an anti-CTLA-4 mAb tremelimumab, or durvalumab and tremelimumab combination in patients with PD-L1-low/negative R/M HNSCC (TC < 25%). ORR of the durvalumab monotherapy arm (n = 67) was 9.2%. ORR for patients with TC < 1% and TC < 10% were 8.8% and 8.9%, respectively [31].
The following randomized phase III study (EAGLE) tested durvalumab vs. durvalumab plus tremelimumab vs. standard chemotherapy in patients with advanced HNSCC regardless of baseline PD-L1 status [32]. In the total population (n = 240), durvalumab monotherapy achieved an ORR of 17.9% and a mOS of 7.6 months, which was not significantly different from the standard control arm (HR 1.04, p = 0.76). Median OS in patients with PD-L1 TC ≥ 25% (n = 68) and TC < 25% (n = 172) were 9.8 months and 7.6 months, respectively.

3.3. Challenges and Limitations of PD-L1 as a Predictive Marker

While PD-L1 expression is widely used as a predictive marker to immune checkpoint therapy and is required in certain indications, several challenges and limitations exist. First, several assays utilizing different antibodies for PD-L1 detection are being used and these tests do not always reproduce the same value. When analyzing the concordance between various PD-L1 assays in the HNSCC population, substantial differences in the level of positivity between assays were observed [36]. With percentages of positive immune and tumor cells varying greatly between assays, the eligibility for certain checkpoint inhibitor regimens was dependent on the choice of the assay [37]. Compounded with the fact that there are varying cut-offs determining PD-L1 positivity (>1% and >50%) and a plethora of PD-L1 detection IHC antibodies utilized with unknown comparative performance characteristics, the inconsistencies of these components complicate PD-L1 testing [38]. This begs consideration for other techniques that can more effectively couple the studies of multiple immune markers. In the setting of non-small cell lung cancer, a multicenter study noted there to be strong concordance between 3 of 4 tested assays (28-8, 22C3, and SP263) when scoring tumor cells [39,40,41,42,43]. Although these assays had significantly less concordance when scoring immune cells, it shows promise for future studies identifying more accurate assays that are able to establish a more standardized diagnosis, especially among HNSCC.
Secondly, PD-L1 expression is highly dynamic and can change significantly over time and by intervening therapies [44]. The data on the effect of chemotherapy on PD-L1 expression is mixed. In NSCLC, while some studies showed that chemotherapy given prior to surgery was shown to increase PD-L1 expression, other studies demonstrated the opposite effect of chemotherapy on PD-L1 expression [45,46]. Additionally, chemotherapy may have distinct effects on tumor cells and immune cells, and different chemotherapeutic agents may have different effects [47]. In HNSCC, Oak et al. demonstrated the cisplatin-based chemotherapy upregulated tumor PD-L1 expression. The data on the effects of the immune checkpoint therapies on the PD-L1 expression is lacking. In an autopsy study, a decrease in PD-L1 expression was observed after pembrolizumab treatment in patients with NSCLC [48].
The current guideline does not specify the timing of the tissue to be tested for PD-L1 or take into consideration any intervening systemic or radiation therapy. This may also potentially explain why some PD-L1-negative patients still benefit from therapy or vice versa, as the immune composition of their tumor microenvironment may have fluctuated since the time of biopsy and the location of the tissue acquisition. Furthermore, it is important to note that HNSCC forms dynamic tumors manifesting high levels of inter- and intra-tumor heterogeneity that has led to significant disparities in therapeutic response to the same treatments [49]. Intra-tumor heterogeneity and intertumoral heterogeneity between primary tumors and nodal metastases were prevalent as well, being seen in 53% of cases [50]. The molecular diversity and continued mutations of HNSCC tumors also make it difficult to obtain truly representative tissue biopsies [51]. Specifically, it can be difficult to determine variations prior to and after medical interventions, as well as during surgical inventions in which it is imperative to identify the tumor nest, stroma, and margin of invasion [52].
There is also a lack of standardization based on the location of the biopsy, whether it is a primary, nodal, or distant metastatic lesion. Across several cancers, PD-L1 expression varies across primary and metastatic lesions [53]. This suggests a difference in the immune microenvironment across sites of metastases; however, there are no clear guidelines regarding PD-L1 positivity depending on tissue biopsied. Standardizing the timing and anatomic location of PD-L1 testing can help to remedy many of the discrepancies that exist with immune assays of biopsied HNSCC tissues. In addition, it’s important to biopsy larger samples of tissue, as up to 35% of small volume biopsies were misclassified resulting in false-negative and false-positive results [54].

4. Other Biomarkers in HNSCC

The tumor mutational burden (TMB), defined as the total number of mutations found in cancer cells, has been considered as a potential biomarker in assessing ICI therapy [55]. Generally, an increased TMB results in the production and expression of more neo-antigens on MHC proteins that can be recognized by T cells [56]. In a retrospective study analyzing 12 clinical trials including over 1770 patients with solid tumors (including 235 HNSCC patients) treated with pembrolizumab, a high mutational burden (with a cut-off value of 175 mutations/exome) was associated with improved ORR (p = 0.016), PFS (p < 0.005), and OS (p = 0.029), independent of PD-L1 expression [57].
Similarly, the quantity of neoantigen has been explored for its predictive value for ICI response. Cancer neoantigens are aberrant antigens produced by cancer-specific genetic alterations such as mutation, alternative splicing, and gene arrangement as well as tumor-specific viral genes. Immunogenic neoantigens are determined based on the affinity of putative neoantigens for the patient-specific HLA class I molecules using prediction algorithms. High neoantigen load has been associated with a high level of tumor-infiltrating immune cells and high expression of pro-immune gene expression in various solid tumors [58,59]. In melanoma patients, neoantigen load was found to be significantly associated with the clinical benefit of ipilimumab [60]. Such predictive value of neoantigen load has been reported in other tumor types including urothelial carcinoma, renal cell carcinoma, and lung cancer [61,62,63,64]. In HNSCC, in addition to single nucleotide variation-derived neoantigens, a high level of frameshift insertion-and deletion-derived neoantigens and HPV/EBV viral antigens may generate anti-tumor immune responses [65]. Hanna et al. reported a higher frequency of frameshift events in ICI responders compared to non-responders in patients with HNSCC [66]. Immune cell infiltration into the tumor microenvironment has also shown prognostic implications [67,68]. Analysis of the levels of CD4+ helper T cells, CD8+ cytotoxic T cells, and FoxP3+ regulatory T cells demonstrated an association of higher tumor-infiltrating lymphocytes (TILs) with improved OS and disease-specific survival even after controlling for other variables. Furthermore, CD4+ cell-rich tumors were better candidates for treatment and associated with higher OS rates than CD4+ cell-depleted tumors [69]. It was also shown that there was a positive correlation between CD3+ T cell infiltration and favorable clinical outcomes in the treatment of HNSCC tumors [70]. Recent studies unveiled the role of tissue-resident memory CD8+T (Trm) cells in anti-tumor immunity. These CD103+CD8+ Trm cells play roles in protective immunity. In human cancers, tumor-infiltration Trm cells have been found to promote anti-tumor immunity and are associated with improved survival in HNSCC [71,72]. Trm cells in tumors are highly enriched with immune checkpoints including PD-L1 and LAG3 and are expanded significantly early with ICI therapy and the levels of CD8+CD103+ TRM are associated with improved patient survival suggesting a critical role Trm in ICI-mediated immune response and its potential predictive value [71,73]. In addition to tumor-infiltrating immune cell composition, the expression of specific immune-related genes in TME has been shown to correlate with clinical outcomes with ICIs in the HNSCC population. Analysis of IFN-γ gene signature of 18 genes (T-cell-inflamed gene expression profile, TcellinfGEP) in HNSCC patient tissues from KEYNOTE-012 study showed independent predictive values of TMB and GEP score with ORR to pembrolizumab therapy response in HPV and EBV negative HNSCC. Interestingly, only the GEP score showed a correlation with pembrolizumab response in HPV- or EBV-mediated tumors [74]. A single-institution retrospective analysis of genomic and transcriptomic data of patients with HNSCC treated with anti-PD-1/PD-L1 ICI showed a correlation between somatic frameshift events and objective responses (p = 0.03) [75].
As the tissue-based biomarker is not always feasible and as in PD-L1 expression there is significant inherent heterogeneity in tumor tissue, blood-based biomarkers have been explored as an alternative predictive marker to tissue-based ones. Baseline and on-treatment dynamics of peripheral immune cell numbers and/or ratios were examined for potential predictive values. A greater pre-treatment neutrophil-lymphocyte ratio (NLR) is associated with poorer overall rates of survival [76]. A meta-analysis of 6479 patients further corroborates this as combined HR for OS in patients with elevated NLR was 1.78 (95% CI 1.53–2.07; p < 0.001) relative to patients with low to normal NLR [77]. NLR has also been shown to exhibit a predictive value for anti-PD-1 therapy response, as low NLR (<6.2) 6 weeks into anti-PD-1 therapy was associated with longer PFS (8.7 vs. 2.9 months, p = 0.001) [77]. Circulating tumor DNA-based TMB (bTMB) was evaluated as a predictive marker for immunotherapy in advanced HNSCC. In the randomized phase 3 EAGLE study where R/M HNSCC patients received chemotherapy, durvalumab, or durvalumab and tremelimumab, a higher bTMB was associated with a greater survival benefit of ICIs over chemotherapy [78].

5. Novel Biomarkers and Future Directions

There are several emerging techniques being developed and utilized in order to identify reliable, reproducible, and clinically relevant biomarkers for early diagnosis, detection, and development of novel therapeutic strategies (Figure 1). Currently, there are limited biomarkers and few targeted approaches for the treatment of advanced HNSCC [79]. There is a need to utilize recent advances including single-cell sequencing approaches, spatial transcriptomics, epigenetic technologies including single-cell ATAC-sequencing to study the transcriptome that governs intra-tumor heterogeneity in HNSCC [80]. These high-resolution strategies will help in delineating the complex tumor–immune interaction occurring within the tumor immune microenvironment [81]. This ultimately allows for the stratification of distinct cellular subpopulations within a tumor in order to better define the immune status of a tumor.
Another strategy to consider in studying and stratifying tumors is DNA and histone methylation analysis. Antigen Processing Machinery (APM) components and IFN-γ stimulated genes (ISG) are transcriptionally silenced in tumors through epigenetic silencing mediated by DNA or histone methylation [82,83]. Therefore, using DNA and histone methylation analysis to identify the transcriptional state of these APM and ISG genes will also help in predicting patient’s response to immunotherapy, with the transcriptional status of these genes defining tumor immune status as being either HOT or COLD. Epigenomic diagnostics have shown DNA methylation of various prognostic transcription factors and immune checkpoints (PITX2, SHOX2, SEPT9, PD-1, and CTLA4) to be correlated with HNSCC and potentially associated with targeted therapies [84]. Clinical assays should be developed to monitor the DNA methylation status utilizing patient DNA. Depending on the epigenetic state, DNMT inhibitors or the recently FDA-approved drug, tazemetostat could be better utilized in the clinic in combination with immunotherapy and increase overall survival (Figure 1A) [85].
Additionally, various studies of independent prognostic-related proteins (IPP) and IPP signatures are being conducted in order to identify proteomic signatures that can function as prognostic markers that guide management and provide targeted therapy options [86,87]. More recently, YAP1, a downstream transcriptional component of the Hippo pathway, has been shown to contribute to resistance mechanisms in HNSCC [88]. FAT1, the upstream negative regulator of YAP1 is mutated in 30% of HNSCC. Normally, FAT1 through a cascade of signaling events phosphorylates YAP1, thereby retaining it in the cytoplasm and inhibiting the oncogenic transcriptional program [89]. FAT1 mutation or phosphorylation of YAP1 could be utilized as a biomarker for predicting the outcome, disease recurrence, and response to trametinib in patients (Figure 1B) [90]. Metabolomic markers could be integrated with epigenomic and proteomic markers and could be the focus for the future to develop novel biomarkers in HNSCC. Oncometabolites such as acylcarnitine and 2-hydroxyglutarate have been identified as potentially non-invasive biomarkers; however, studies validating the clinical efficacy of these targets must be conducted to validate these findings and have a clinical application [91,92].
Plasma-based diagnostic markers have also grown to become an area of emphasis, as this would function as a virtually non-invasive means of diagnosis or treatment monitoring of HNSCC [93]. Plasma Melanoma-Antigen (MLANA) recognized by T-cells has been identified as an effective plasma-based biomarker in monitoring HNSCC patients undergoing chemoradiation therapy [94]. Investigation of extracellular vesicles, exosomes, containing potential biomarkers has indicated the potential to guide future therapies and establish more sensitive monitoring of existing therapies [95]. These exosomes have been shown to carry PDL1, which is upregulated by IFN-γ and suppresses CD8+ T cell function, and facilitates tumor growth without direct expression on the tumor surface [96].
Various tumor- and immune cell-derived immune factors collectively determine the tumor immune microenvironment (TIME) characteristics. Among several cytokine and cytokine receptors, the colony-stimulating factor-1 (CSF-1) secreted by tumor cells interacts with various immune cells in the environment, particularly tumor-associated macrophages (TAM). Such interaction induces TAM into immune suppressive M2 types promoting immune suppression and tumor progression [97]. A high level of M2 TAM in the tumor microenvironment is associated with poor prognosis in various tumors including HNSCC and also resistance to ICIs [98,99,100,101]. CSF-1/CSF1-targeting investigational agents are being evaluated alone or in combination with ICI [102,103].
Additionally, the use of specialized tissue models that better mimic native tumor microenvironments, including the immune signatures and intratumor heterogeneity, allow for more accurate testing of targeted therapies [104]. A relatively new tissue model that has the potential to revolutionize the way cancers are studied and, in turn, treated, is the organoid model. An organoid model is a 3D, multicellular in vitro tissue construct that mimics a corresponding in vivo organ and can be utilized to study aspects of physiology and pathologies associated with that organ [105]. Organoids have been shown to be able to express the diversity of carcinoma subtypes and are capable of predicting in vivo drug sensitivity better than other models [106]. In the case of HNSCC, organoids have the potential to open the door to entirely new subsets of immune markers for targeted therapy, as drug screens have shown sensitivity to targeted drugs that have not been traditionally used in treating HNSCC (Figure 1C) [107]. In conclusion, the development of potential novel biomarkers and accompanying sensitive clinical diagnostic testing tools is important to design better treatment strategies. This will allow for the development of targeted immunotherapies that alleviate the morbidity and mortality associated with HNSCC.

Author Contributions

J.C.P.: project administration, writing—review and editing, H.N.K.: writing—original draft preparation, S.V.S.: supervision, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and Neck Squamous Cell Carcinoma. Nat. Rev. Dis. Primer 2020, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  2. Shaw, R.; Beasley, N. Aetiology and Risk Factors for Head and Neck Cancer: United Kingdom National Multidisciplinary Guidelines. J. Laryngol. Otol. 2016, 130, S9–S12. [Google Scholar] [CrossRef]
  3. Pisani, P.; Airoldi, M.; Allais, A.; Aluffi Valletti, P.; Battista, M.; Benazzo, M.; Briatore, R.; Cacciola, S.; Cocuzza, S.; Colombo, A.; et al. Metastatic Disease in Head & Neck Oncology. Acta Otorhinolaryngol. Ital. 2020, 40, S1–S86. [Google Scholar] [CrossRef]
  4. Lee, Y.-G.; Kang, E.J.; Keam, B.; Choi, J.-H.; Kim, J.-S.; Park, K.U.; Lee, K.E.; Kwon, J.H.; Lee, K.-W.; Kim, M.K.; et al. Treatment Strategy and Outcomes in Locally Advanced Head and Neck Squamous Cell Carcinoma: A Nationwide Retrospective Cohort Study (KCSG HN13–01). BMC Cancer 2020, 20, 813. [Google Scholar] [CrossRef]
  5. Ribeiro, I.P.; Caramelo, F.; Esteves, L.; Menoita, J.; Marques, F.; Barroso, L.; Miguéis, J.; Melo, J.B.; Carreira, I.M. Genomic Predictive Model for Recurrence and Metastasis Development in Head and Neck Squamous Cell Carcinoma Patients. Sci. Rep. 2017, 7, 13897. [Google Scholar] [CrossRef] [Green Version]
  6. Larkins, E.; Blumenthal, G.M.; Yuan, W.; He, K.; Sridhara, R.; Subramaniam, S.; Zhao, H.; Liu, C.; Yu, J.; Goldberg, K.B.; et al. FDA Approval Summary: Pembrolizumab for the Treatment of Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma with Disease Progression on or after Platinum-Containing Chemotherapy. Oncologist 2017, 22, 873–878. [Google Scholar] [CrossRef] [Green Version]
  7. Pei, R.; Shi, Y.; Lv, S.; Dai, T.; Zhang, F.; Liu, S.; Wu, B. Nivolumab vs. Pembrolizumab for Treatment of US Patients with Platinum-Refractory Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: A Network Meta-Analysis and Cost-Effectiveness Analysis. JAMA Netw. Open 2021, 4, e218065. [Google Scholar] [CrossRef]
  8. Prasad, V.; Kaestner, V. Nivolumab and Pembrolizumab: Monoclonal Antibodies against Programmed Cell Death-1 (PD-1) That Are Interchangeable. Semin. Oncol. 2017, 44, 132–135. [Google Scholar] [CrossRef]
  9. Pai, S.I.; Faivre, S.; Licitra, L.; Machiels, J.-P.; Vermorken, J.B.; Bruzzi, P.; Gruenwald, V.; Giglio, R.E.; Leemans, C.R.; Seiwert, T.Y.; et al. Comparative Analysis of the Phase III Clinical Trials of Anti-PD1 Monotherapy in Head and Neck Squamous Cell Carcinoma Patients (CheckMate 141 and KEYNOTE 040). J. Immunother. Cancer 2019, 7, 96. [Google Scholar] [CrossRef]
  10. Gillison, M.L.; Blumenschein, G.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.J.; Kasper, S.; Vokes, E.E.; Even, C.; et al. CheckMate 141: 1-Year Update and Subgroup Analysis of Nivolumab as First-Line Therapy in Patients with Recurrent/Metastatic Head and Neck Cancer. Oncologist 2018, 23, 1079–1082. [Google Scholar] [CrossRef] [Green Version]
  11. Cohen, E.E.W.; Bell, R.B.; Bifulco, C.B.; Burtness, B.; Gillison, M.L.; Harrington, K.J.; Le, Q.-T.; Lee, N.Y.; Leidner, R.; Lewis, R.L.; et al. The Society for Immunotherapy of Cancer Consensus Statement on Immunotherapy for the Treatment of Squamous Cell Carcinoma of the Head and Neck (HNSCC). J. Immunother. Cancer 2019, 7, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bauml, J.M.; Aggarwal, C.; Cohen, R.B. Immunotherapy for Head and Neck Cancer: Where Are We Now and Where Are We Going? Ann. Transl. Med. 2019, 7, S75. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, H.; Kwon, M.; Kim, B.; Jung, H.A.; Sun, J.-M.; Lee, S.-H.; Park, K.; Ahn, M.-J. Clinical Outcomes of Immune Checkpoint Inhibitors for Patients with Recurrent or Metastatic Head and Neck Cancer: Real-World Data in Korea. BMC Cancer 2020, 20, 727. [Google Scholar] [CrossRef]
  14. Xu, K.; Fu, Y.; Han, Y.; Xia, R.; Xu, S.; Duan, S.; Zhang, Z.; Li, J. Fewer Tumour-Specific PD-1+CD8+ TILs in High-Risk “Infiltrating” HPV− HNSCC. Br. J. Cancer 2020, 123, 932–941. [Google Scholar] [CrossRef]
  15. Yi, M.; Niu, M.; Xu, L.; Luo, S.; Wu, K. Regulation of PD-L1 Expression in the Tumor Microenvironment. J. Hematol. Oncol. 2021, 14, 10. [Google Scholar] [CrossRef]
  16. Oliva, M.; Spreafico, A.; Taberna, M.; Alemany, L.; Coburn, B.; Mesia, R.; Siu, L.L. Immune Biomarkers of Response to Immune-Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma. Ann. Oncol. 2019, 30, 57–67. [Google Scholar] [CrossRef]
  17. Kim, H.R.; Ha, S.-J.; Hong, M.H.; Heo, S.J.; Koh, Y.W.; Choi, E.C.; Kim, E.K.; Pyo, K.H.; Jung, I.; Seo, D.; et al. PD-L1 Expression on Immune Cells, but Not on Tumor Cells, Is a Favorable Prognostic Factor for Head and Neck Cancer Patients. Sci. Rep. 2016, 6, 36956. [Google Scholar] [CrossRef] [Green Version]
  18. Kulangara, K.; Hanks, D.A.; Waldroup, S.; Peltz, L.; Shah, S.; Roach, C.; Juco, J.W.; Emancipator, K.; Stanforth, D. Development of the Combined Positive Score (CPS) for the Evaluation of PD-L1 in Solid Tumors with the Immunohistochemistry Assay PD-L1 IHC 22C3 PharmDx. J. Clin. Oncol. 2017, 35, e14589. [Google Scholar] [CrossRef]
  19. Mehra, R.; Seiwert, T.Y.; Gupta, S.; Weiss, J.; Gluck, I.; Eder, J.P.; Burtness, B.; Tahara, M.; Keam, B.; Kang, H.; 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, 153–159. [Google Scholar] [CrossRef]
  20. Chow, L.Q.M.; Haddad, R.; Gupta, S.; Mahipal, A.; Mehra, R.; Tahara, M.; Berger, R.; Eder, J.P.; Burtness, B.; Lee, S.-H.; 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, 3838–3845. [Google Scholar] [CrossRef]
  21. Haddad, R.; Seiwert, T.; Pfister, D.G.; Worden, F.; Liu, S.V.; Gilbert, J.; Saba, N.F.; Weiss, J.; Wirth, L.J.; Sukari, A.; et al. Pembrolizumab after Progression on Platinum and Cetuximab in Head and Neck Squamous Cell Carcinoma (HNSCC): Results from KEYNOTE-055. Ann. Oncol. 2016, 27, vi330. [Google Scholar] [CrossRef]
  22. Bauml, J.; Seiwert, T.Y.; Pfister, D.G.; Worden, F.; Liu, S.V.; Gilbert, J.; Saba, N.F.; Weiss, J.; Wirth, L.; Sukari, A.; 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, 1542–1549. [Google Scholar] [CrossRef] [PubMed]
  23. Cohen, E.E.W.; Machiels, J.-P.H.; Harrington, K.J.; Burtness, B.; Shin, S.W.; Gause, C.K.; Swift, A.M.; Brown, H.; Perrone, A.M.; Cheng, J.D.; et al. KEYNOTE-040: A Phase III Randomized Trial of Pembrolizumab (MK-3475) versus Standard Treatment in Patients with Recurrent or Metastatic Head and Neck Cancer. J. Clin. Oncol. 2015, 33, TPS6084. [Google Scholar] [CrossRef]
  24. Harrington, K.J.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.F.; Ahn, M.-J.; Soria, A.; Machiels, J.-P.H.; Mach, N.; Mehra, R.; et al. Quality of Life with Pembrolizumab for Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma: KEYNOTE-040. J. Natl. Cancer Inst. 2021, 113, 171–181. [Google Scholar] [CrossRef] [PubMed]
  25. Cohen, E.E.W.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.-J.; Soria, A.; Machiels, J.-P.; Mach, N.; Mehra, R.; et al. 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, 156–167. [Google Scholar] [CrossRef]
  26. Ferris, R.L.; Blumenschein, G.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.J.; Kasper, S.; Vokes, E.E.; Even, C.; 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. [Google Scholar] [CrossRef]
  27. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, Å.; et al. Pembrolizumab Alone or with Chemotherapy versus Cetuximab with Chemotherapy for Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-048): A Randomised, Open-Label, Phase 3 Study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  28. Beyaert, S.; Machiels, J.-P.; Schmitz, S. Vaccine-Based Immunotherapy for Head and Neck Cancers. Cancers 2021, 13, 6041. [Google Scholar] [CrossRef]
  29. Kwok, G.; Yau, T.C.C.; Chiu, J.W.; Tse, E.; Kwong, Y.-L. Pembrolizumab (Keytruda). Hum. Vaccines Immunother. 2016, 12, 2777–2789. [Google Scholar] [CrossRef] [Green Version]
  30. Zandberg, D.P.; Algazi, A.P.; Jimeno, A.; Good, J.S.; Fayette, J.; Bouganim, N.; Ready, N.E.; Clement, P.M.; Even, C.; Jang, R.W.; 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. [Google Scholar] [CrossRef] [Green Version]
  31. Siu, L.L.; Even, C.; Mesía, R.; Remenar, E.; Daste, A.; Delord, J.-P.; Krauss, J.; Saba, N.F.; Nabell, L.; Ready, N.E.; 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, 195–203. [Google Scholar] [CrossRef] [PubMed]
  32. Ferris, R.L.; Haddad, R.; Even, C.; Tahara, M.; Dvorkin, M.; Ciuleanu, T.E.; Clement, P.M.; Mesia, R.; Kutukova, S.; Zholudeva, L.; et al. Durvalumab with or without Tremelimumab in Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: EAGLE, a Randomized, Open-Label Phase III Study. Ann. Oncol. 2020, 31, 942–950. [Google Scholar] [CrossRef] [PubMed]
  33. Colevas, A.D.; Bahleda, R.; Braiteh, F.; Balmanoukian, A.; Brana, I.; Chau, N.G.; Sarkar, I.; Molinero, L.; Grossman, W.; Kabbinavar, F.; et al. Safety and Clinical Activity of Atezolizumab in Head and Neck Cancer: Results from a Phase I Trial. Ann. Oncol. 2018, 29, 2247–2253. [Google Scholar] [CrossRef] [PubMed]
  34. Rischin, D.; Harrington, K.J.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G.; Psyrri, A.; Baste, N.; Neupane, P.C.; Bratland, A.; et al. Protocol-Specified Final Analysis of the Phase 3 KEYNOTE-048 Trial of Pembrolizumab (Pembro) as First-Line Therapy for Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma (R/M HNSCC). J. Clin. Oncol. 2019, 37, 6000. [Google Scholar] [CrossRef]
  35. Segal, N.H.; Ou, S.-H.I.; Balmanoukian, A.; Fury, M.G.; Massarelli, E.; Brahmer, J.R.; Weiss, J.; Schöffski, P.; Antonia, S.J.; Massard, C.; et al. Safety and Efficacy of Durvalumab in Patients with Head and Neck Squamous Cell Carcinoma: Results from a Phase I/II Expansion Cohort. Eur. J. Cancer 2019, 109, 154–161. [Google Scholar] [CrossRef] [PubMed]
  36. Girolami, I.; Pantanowitz, L.; Barberis, M.; Paolino, G.; Brunelli, M.; Vigliar, E.; Munari, E.; Satturwar, S.; Troncone, G.; Eccher, A. Challenges Facing Pathologists Evaluating PD-L1 in Head & Neck Squamous Cell Carcinoma. J. Oral Pathol. Med. 2021, 50, 864–873. [Google Scholar] [CrossRef]
  37. Ribbat-Idel, J.; Dressler, F.F.; Krupar, R.; Watermann, C.; Paulsen, F.-O.; Kuppler, P.; Klapper, L.; Offermann, A.; Wollenberg, B.; Rades, D.; et al. Performance of Different Diagnostic PD-L1 Clones in Head and Neck Squamous Cell Carcinoma. Front. Med. 2021, 8, 640515. [Google Scholar] [CrossRef]
  38. Martinez Marti, A.; Martinez, P.; Navarro, A.; Cedres, S.; Murtra-Garrell, N.; Salva, F.; Sansano, I.; Romero, L.; Felip, E.; Nuciforo, P. Concordance of PD-L1 Expression by Different Immunohistochemistry (IHC) Definitions and in Situ Hybridization (ISH) in Squamous Cell Carcinoma (SCC) of the Lung. J. Clin. Oncol. 2014, 32, 7569. [Google Scholar] [CrossRef]
  39. Torlakovic, E.; Lim, H.J.; Adam, J.; Barnes, P.; Bigras, G.; Chan, A.W.H.; Cheung, C.C.; Chung, J.-H.; Couture, C.; Fiset, P.O.; et al. “Interchangeability” of PD-L1 Immunohistochemistry Assays: A Meta-Analysis of Diagnostic Accuracy. Mod. Pathol. 2020, 33, 4–17. [Google Scholar] [CrossRef]
  40. Hendry, S.; Byrne, D.J.; Wright, G.M.; Young, R.J.; Sturrock, S.; Cooper, W.A.; Fox, S.B. Comparison of Four PD-L1 Immunohistochemical Assays in Lung Cancer. J. Thorac. Oncol. 2018, 13, 367–376. [Google Scholar] [CrossRef] [Green Version]
  41. Prince, E.A.; Sanzari, J.K.; Pandya, D.; Huron, D.; Edwards, R. Analytical Concordance of PD-L1 Assays Utilizing Antibodies from FDA-Approved Diagnostics in Advanced Cancers: A Systematic Literature Review. JCO Precis. Oncol. 2021, 953–973. [Google Scholar] [CrossRef] [PubMed]
  42. Rimm, D.L.; Han, G.; Taube, J.M.; Yi, E.S.; Bridge, J.A.; Flieder, D.B.; Homer, R.; West, W.W.; Wu, H.; Roden, A.C.; et al. A Prospective, Multi-Institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non–Small Cell Lung Cancer. JAMA Oncol. 2017, 3, 1051. [Google Scholar] [CrossRef] [PubMed]
  43. Adam, J.; Le Stang, N.; Rouquette, I.; Cazes, A.; Badoual, C.; Pinot-Roussel, H.; Tixier, L.; Danel, C.; Damiola, F.; Damotte, D.; et al. Multicenter Harmonization Study for PD-L1 IHC Testing in Non-Small-Cell Lung Cancer. Ann. Oncol. 2018, 29, 953–958. [Google Scholar] [CrossRef] [PubMed]
  44. Tang, J.; Yu, J.X.; Hubbard-Lucey, V.M.; Neftelinov, S.T.; Hodge, J.P.; Lin, Y. The Clinical Trial Landscape for PD1/PDL1 Immune Checkpoint Inhibitors. Nat. Rev. Drug Discov. 2018, 17, 854–855. [Google Scholar] [CrossRef]
  45. Guo, L.; Song, P.; Xue, X.; Guo, C.; Han, L.; Fang, Q.; Ying, J.; Gao, S.; Li, W. Variation of Programmed Death Ligand 1 Expression after Platinum-Based Neoadjuvant Chemotherapy in Lung Cancer. J. Immunother. 2019, 42, 215–220. [Google Scholar] [CrossRef]
  46. Sheng, J.; Fang, W.; Yu, J.; Chen, N.; Zhan, J.; Ma, Y.; Yang, Y.; Huang, Y.; Huang, Y.; Zhao, H.; et al. Expression of Programmed Death Ligand-1 on Tumor Cells Varies Pre and Post Chemotherapy in Non-Small Cell Lung Cancer. Sci. Rep. 2016, 6, 20090. [Google Scholar] [CrossRef] [Green Version]
  47. Rojkó, L.; Reiniger, L.; Téglási, V.; Fábián, K.; Pipek, O.; Vágvölgyi, A.; Agócs, L.; Fillinger, J.; Kajdácsi, Z.; Tímár, J.; et al. Chemotherapy Treatment Is Associated with Altered PD-L1 Expression in Lung Cancer Patients. J. Cancer Res. Clin. Oncol. 2018, 144, 1219–1226. [Google Scholar] [CrossRef]
  48. Takahashi, T.; Tateishi, A.; Bychkov, A.; Fukuoka, J. Remarkable Alteration of PD-L1 Expression after Immune Checkpoint Therapy in Patients with Non-Small-Cell Lung Cancer: Two Autopsy Case Reports. Int. J. Mol. Sci. 2019, 20, 2578. [Google Scholar] [CrossRef] [Green Version]
  49. Canning, M.; Guo, G.; Yu, M.; Myint, C.; Groves, M.W.; Byrd, J.K.; Cui, Y. Heterogeneity of the Head and Neck Squamous Cell Carcinoma Immune Landscape and Its Impact on Immunotherapy. Front. Cell Dev. Biol. 2019, 7, 52. [Google Scholar] [CrossRef]
  50. Haragan, A.; Field, J.K.; Davies, M.P.A.; Escriu, C.; Gruver, A.; Gosney, J.R. Heterogeneity of PD-L1 Expression in Non-Small Cell Lung Cancer: Implications for Specimen Sampling in Predicting Treatment Response. Lung Cancer 2019, 134, 79–84. [Google Scholar] [CrossRef] [Green Version]
  51. Suda, K.; Mitsudomi, T. Inter-Tumor Heterogeneity of PD-L1 Status: Is It Important in Clinical Decision Making? J. Thorac. Dis. 2020, 12, 1770–1775. [Google Scholar] [CrossRef] [PubMed]
  52. Economopoulou, P.; Kotsantis, I.; Kyrodimos, E.; Lianidou, E.S.; Psyrri, A. Liquid Biopsy: An Emerging Prognostic and Predictive Tool in Head and Neck Squamous Cell Carcinoma (HNSCC). Focus on Circulating Tumor Cells (CTCs). Oral Oncol. 2017, 74, 83–89. [Google Scholar] [CrossRef] [PubMed]
  53. Rozenblit, M.; Huang, R.; Danziger, N.; Hegde, P.; Alexander, B.; Ramkissoon, S.; Blenman, K.; Ross, J.S.; Rimm, D.L.; Pusztai, L. Comparison of PD-L1 Protein Expression between Primary Tumors and Metastatic Lesions in Triple Negative Breast Cancers. J. Immunother. Cancer 2020, 8, e001558. [Google Scholar] [CrossRef] [PubMed]
  54. Bigras, G.; Mairs, S.; Swanson, P.E.; Morel, D.; Lai, R.; Izevbaye, I. Small Biopsies Misclassify up to 35% of PD-L1 Assessments in Advanced Lung Non–Small Cell Lung Carcinomas. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 701–708. [Google Scholar] [CrossRef] [PubMed]
  55. Ritterhouse, L.L. Tumor Mutational Burden. Cancer Cytopathol. 2019, 127, 735–736. [Google Scholar] [CrossRef]
  56. Jardim, D.L.; Goodman, A.; de Melo Gagliato, D.; Kurzrock, R. The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. Cancer Cell 2021, 39, 154–173. [Google Scholar] [CrossRef]
  57. Cristescu, R.; Aurora-Garg, D.; Albright, A.; Xu, L.; Liu, X.Q.; Loboda, A.; Lang, L.; Jin, F.; Rubin, E.H.; Snyder, A.; et al. Tumor Mutational Burden Predicts the Efficacy of Pembrolizumab Monotherapy: A Pan-Tumor Retrospective Analysis of Participants with Advanced Solid Tumors. J. Immunother. Cancer 2022, 10, e003091. [Google Scholar] [CrossRef]
  58. Strickland, K.C.; Howitt, B.E.; Shukla, S.A.; Rodig, S.; Ritterhouse, L.L.; Liu, J.F.; Garber, J.E.; Chowdhury, D.; Wu, C.J.; D’Andrea, A.D.; et al. Association and Prognostic Significance of BRCA1/2-Mutation Status with Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes and Expression of PD-1/PD-L1 in High Grade Serous Ovarian Cancer. Oncotarget 2016, 7, 13587–13598. [Google Scholar] [CrossRef] [Green Version]
  59. Zhu, Y.; Meng, X.; Ruan, X.; Lu, X.; Yan, F.; Wang, F. Characterization of Neoantigen Load Subgroups in Gynecologic and Breast Cancers. Front. Bioeng. Biotechnol. 2020, 8, 702. [Google Scholar] [CrossRef]
  60. Van Allen, E.M.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Geukes Foppen, M.H.; Goldinger, S.M.; et al. Genomic Correlates of Response to CTLA-4 Blockade in Metastatic Melanoma. Science 2015, 350, 207–211. [Google Scholar] [CrossRef] [Green Version]
  61. Lazdun, Y.; Si, H.; Creasy, T.; Ranade, K.; Higgs, B.W.; Streicher, K.; Durham, N.M. A New Pipeline to Predict and Confirm Tumor Neoantigens Predict Better Response to Immune Checkpoint Blockade. Mol. Cancer Res. MCR 2021, 19, 498–506. [Google Scholar] [CrossRef]
  62. Matsushita, H.; Sato, Y.; Karasaki, T.; Nakagawa, T.; Kume, H.; Ogawa, S.; Homma, Y.; Kakimi, K. Neoantigen Load, Antigen Presentation Machinery, and Immune Signatures Determine Prognosis in Clear Cell Renal Cell Carcinoma. Cancer Immunol. Res. 2016, 4, 463–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer Immunology. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non-Small Cell Lung Cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal Neoantigens Elicit T Cell Immunoreactivity and Sensitivity to Immune Checkpoint Blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-Deletion-Derived Tumour-Specific Neoantigens and the Immunogenic Phenotype: A Pan-Cancer Analysis. Lancet Oncol. 2017, 18, 1009–1021. [Google Scholar] [CrossRef] [Green Version]
  66. Hanna, G.J.; Lizotte, P.; Cavanaugh, M.; Kuo, F.C.; Shivdasani, P.; Frieden, A.; Chau, N.G.; Schoenfeld, J.D.; Lorch, J.H.; Uppaluri, R.; et al. Frameshift Events Predict Anti-PD-1/L1 Response in Head and Neck Cancer. JCI Insight 2018, 3, 98811. [Google Scholar] [CrossRef] [Green Version]
  67. dos Santos, L.V.; Abrahão, C.M.; William, W.N. Overcoming Resistance to Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinomas. Front. Oncol. 2021, 11, 596290. [Google Scholar] [CrossRef]
  68. Nguyen, N.; Bellile, E.; Thomas, D.; McHugh, J.; Rozek, L.; Virani, S.; Peterson, L.; Carey, T.E.; Walline, H.; Moyer, J.; et al. Tumor Infiltrating Lymphocytes and Survival in Patients with Head and Neck Squamous Cell Carcinoma: Tumor Infiltrating Lymphocytes. Head Neck 2016, 38, 1074–1084. [Google Scholar] [CrossRef] [Green Version]
  69. Spector, M.E.; Bellile, E.; Amlani, L.; Zarins, K.; Smith, J.; Brenner, J.C.; Rozek, L.; Nguyen, A.; Thomas, D.; McHugh, J.B.; et al. Prognostic Value of Tumor-Infiltrating Lymphocytes in Head and Neck Squamous Cell Carcinoma. JAMA Otolaryngol.–Head Neck Surg. 2019, 145, 1012–1019. [Google Scholar] [CrossRef]
  70. de Ruiter, E.J.; Ooft, M.L.; Devriese, L.A.; Willems, S.M. The Prognostic Role of Tumor Infiltrating T-Lymphocytes in Squamous Cell Carcinoma of the Head and Neck: A Systematic Review and Meta-Analysis. Oncoimmunology 2017, 6, e1356148. [Google Scholar] [CrossRef] [Green Version]
  71. Edwards, J.; Wilmott, J.S.; Madore, J.; Gide, T.N.; Quek, C.; Tasker, A.; Ferguson, A.; Chen, J.; Hewavisenti, R.; Hersey, P.; et al. CD103+ Tumor-Resident CD8+ T Cells Are Associated with Improved Survival in Immunotherapy-Naïve Melanoma Patients and Expand Significantly During Anti-PD-1 Treatment. Clin. Cancer Res. 2018, 24, 3036–3045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hewavisenti, R.; Ferguson, A.; Wang, K.; Jones, D.; Gebhardt, T.; Edwards, J.; Zhang, M.; Britton, W.; Yang, J.; Hong, A.; et al. CD103+ Tumor-Resident CD8+ T Cell Numbers Underlie Improved Patient Survival in Oropharyngeal Squamous Cell Carcinoma. J. Immunother. Cancer 2020, 8, e000452. [Google Scholar] [CrossRef] [PubMed]
  73. Mei, X.; Li, H.; Zhou, X.; Cheng, M.; Cui, K. The Emerging Role of Tissue-Resident Memory CD8+ T Lymphocytes in Human Digestive Tract Cancers. Front. Oncol. 2021, 11, 819505. [Google Scholar] [CrossRef] [PubMed]
  74. Park, J.C.; Durbeck, J.; Clark, J.R. Predictive Value of Peripheral Lymphocyte Counts for Immune Checkpoint Inhibitor Efficacy in Advanced Head and Neck Squamous Cell Carcinoma. Mol. Clin. Oncol. 2020, 13, 87. [Google Scholar] [CrossRef] [PubMed]
  75. Haddad, R.I.; Seiwert, T.Y.; Chow, L.Q.M.; Gupta, S.; Weiss, J.; Gluck, I.; Eder, J.P.; Burtness, B.; Tahara, M.; Keam, B.; et al. Influence of Tumor Mutational Burden, Inflammatory Gene Expression Profile, and PD-L1 Expression on Response to Pembrolizumab in Head and Neck Squamous Cell Carcinoma. J. Immunother. Cancer 2022, 10, e003026. [Google Scholar] [CrossRef]
  76. Yang, L.; Huang, Y.; Zhou, L.; Dai, Y.; Hu, G. High Pretreatment Neutrophil-to-Lymphocyte Ratio as a Predictor of Poor Survival Prognosis in Head and Neck Squamous Cell Carcinoma: Systematic Review and Meta-Analysis. Head Neck 2019, 41, 1525–1535. [Google Scholar] [CrossRef]
  77. Mascarella, M.A.; Mannard, E.; Silva, S.D.; Zeitouni, A. Neutrophil-to-Lymphocyte Ratio in Head and Neck Cancer Prognosis: A Systematic Review and Meta-Analysis. Head Neck 2018, 40, 1091–1100. [Google Scholar] [CrossRef]
  78. Li, W.; Wildsmith, S.; Ye, J.; Si, H.; Morsli, N.; He, P.; Shetty, J.; Yovine, A.J.; Holoweckyj, N.; Raja, R.; et al. Plasma-Based Tumor Mutational Burden (BTMB) as Predictor for Survival in Phase III EAGLE Study: Durvalumab (D) ± Tremelimumab (T) versus Chemotherapy (CT) in Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma (R/M HNSCC) after Platinum Failure. J. Clin. Oncol. 2020, 38, 6511. [Google Scholar] [CrossRef]
  79. Eton, O.; Hsu, E.; Patel, A.; Eton, O.; Cartun, R.; Earle, J.; Mnayer, L.; Yu, P.; Kotowitz, J. 467 Anti-EGFR antibody added to ongoing anti-PD-1 antibody treatment for metastatic cutaneous squamous cell carcinoma of the face: Two case reports. J. ImmunoTherapy Cancer 2020, 8, A284–A286. [Google Scholar]
  80. Jin, Y.; Qin, X. Comprehensive Analysis of Transcriptome Data for Identifying Biomarkers and Therapeutic Targets in Head and Neck Squamous Cell Carcinoma. Ann. Transl. Med. 2020, 8, 282. [Google Scholar] [CrossRef]
  81. Zhang, L.; Li, B.; Peng, Y.; Wu, F.; Li, Q.; Lin, Z.; Xie, S.; Xiao, L.; Lin, X.; Ou, Z.; et al. The Prognostic Value of TMB and the Relationship between TMB and Immune Infiltration in Head and Neck Squamous Cell Carcinoma: A Gene Expression-Based Study. Oral Oncol. 2020, 110, 104943. [Google Scholar] [CrossRef]
  82. Fragale, A.; Romagnoli, G.; Licursi, V.; Buoncervello, M.; Del Vecchio, G.; Giuliani, C.; Parlato, S.; Leone, C.; De Angelis, M.; Canini, I.; et al. Antitumor Effects of Epidrug/IFNα Combination Driven by Modulated Gene Signatures in Both Colorectal Cancer and Dendritic Cells. Cancer Immunol. Res. 2017, 5, 604–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hasim, A.; Abudula, M.; Aimiduo, R.; Ma, J.-Q.; Jiao, Z.; Akula, G.; Wang, T.; Abudula, A. Post-Transcriptional and Epigenetic Regulation of Antigen Processing Machinery (APM) Components and HLA-I in Cervical Cancers from Uighur Women. PLoS ONE 2012, 7, e44952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Digomann, D.; Kurth, I.; Tyutyunnykova, A.; Chen, O.; Löck, S.; Gorodetska, I.; Peitzsch, C.; Skvortsova, I.-I.; Negro, G.; Aschenbrenner, B.; et al. The CD98 Heavy Chain Is a Marker and Regulator of Head and Neck Squamous Cell Carcinoma Radiosensitivity. Clin. Cancer Res. 2019, 25, 3152–3163. [Google Scholar] [CrossRef] [PubMed]
  85. Dan, H.; Zhang, S.; Zhou, Y.; Guan, Q. DNA Methyltransferase Inhibitors: Catalysts for Antitumour Immune Responses. OncoTargets Ther. 2019, 12, 10903–10916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wu, D.; Gong, P.; Zeng, Q.; Zhang, W.; Xie, F.; Zhou, X. Prognostic Implication of Proteomic Profiles in Head and Neck Squamous Cell Carcinoma. Clin. Chim. Acta 2020, 509, 304–309. [Google Scholar] [CrossRef] [PubMed]
  87. Sun, M.; Chen, S.; Fu, M. Model Establishment of Prognostic-Related Immune Genes in Laryngeal Squamous Cell Carcinoma. Medicine 2021, 100, e24263. [Google Scholar] [CrossRef] [PubMed]
  88. Omori, H.; Nishio, M.; Masuda, M.; Miyachi, Y.; Ueda, F.; Nakano, T.; Sato, K.; Mimori, K.; Taguchi, K.; Hikasa, H.; et al. YAP1 Is a Potent Driver of the Onset and Progression of Oral Squamous Cell Carcinoma. Sci. Adv. 2020, 6, eaay3324. [Google Scholar] [CrossRef] [Green Version]
  89. Martin, D.; Degese, M.S.; Vitale-Cross, L.; Iglesias-Bartolome, R.; Valera, J.L.C.; Wang, Z.; Feng, X.; Yeerna, H.; Vadmal, V.; Moroishi, T.; et al. Assembly and Activation of the Hippo Signalome by FAT1 Tumor Suppressor. Nat. Commun. 2018, 9, 2372. [Google Scholar] [CrossRef]
  90. Mudianto, T.; Campbell, K.M.; Webb, J.; Zolkind, P.; Skidmore, Z.L.; Riley, R.; Barnell, E.K.; Ozgenc, I.; Giri, T.; Dunn, G.P.; et al. Yap1 Mediates Trametinib Resistance in Head and Neck Squamous Cell Carcinomas. Clin. Cancer Res. 2021, 27, 2326–2339. [Google Scholar] [CrossRef]
  91. Mukherjee, P.K.; Funchain, P.; Retuerto, M.; Jurevic, R.J.; Fowler, N.; Burkey, B.; Eng, C.; Ghannoum, M.A. Metabolomic Analysis Identifies Differentially Produced Oral Metabolites, Including the Oncometabolite 2-Hydroxyglutarate, in Patients with Head and Neck Squamous Cell Carcinoma. BBA Clin. 2017, 7, 8–15. [Google Scholar] [CrossRef]
  92. Vitório, J.G.; Duarte-Andrade, F.F.; Dos Santos Fontes Pereira, T.; Fonseca, F.P.; Amorim, L.S.D.; Martins-Chaves, R.R.; Gomes, C.C.; Canuto, G.A.B.; Gomez, R.S. Metabolic Landscape of Oral Squamous Cell Carcinoma. Metabolomics 2020, 16, 105. [Google Scholar] [CrossRef] [PubMed]
  93. Mathew, M.; Zade, M.; Mezghani, N.; Patel, R.; Wang, Y.; Momen-Heravi, F. Extracellular Vesicles as Biomarkers in Cancer Immunotherapy. Cancers 2020, 12, 2825. [Google Scholar] [CrossRef] [PubMed]
  94. Rodrigues-Junior, D.M.; Tan, S.S.; Lim, S.K.; de Souza Viana, L.; Carvalho, A.L.; Vettore, A.L.; Iyer, N.G. High Expression of MLANA in the Plasma of Patients with Head and Neck Squamous Cell Carcinoma as a Predictor of Tumor Progression. Head Neck 2019, 41, 1199–1205. [Google Scholar] [CrossRef] [PubMed]
  95. Rodrigues-Junior, D.M.; Tan, S.S.; de Souza Viana, L.; Carvalho, A.L.; Lim, S.K.; Iyer, N.G.; Vettore, A.L. A Preliminary Investigation of Circulating Extracellular Vesicles and Biomarker Discovery Associated with Treatment Response in Head and Neck Squamous Cell Carcinoma. BMC Cancer 2019, 19, 373. [Google Scholar] [CrossRef]
  96. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 Contributes to Immunosuppression and Is Associated with Anti-PD-1 Response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  97. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R Inhibition Alters Macrophage Polarization and Blocks Glioma Progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [Green Version]
  98. Wolf, G.T.; Chepeha, D.B.; Bellile, E.; Nguyen, A.; Thomas, D.; McHugh, J.; University of Michigan Head and Neck SPORE Program. Tumor Infiltrating Lymphocytes (TIL) and Prognosis in Oral Cavity Squamous Carcinoma: A Preliminary Study. Oral Oncol. 2015, 51, 90–95. [Google Scholar] [CrossRef] [Green Version]
  99. Kumar, A.T.; Knops, A.; Swendseid, B.; Martinez-Outschoom, U.; Harshyne, L.; Philp, N.; Rodeck, U.; Luginbuhl, A.; Cognetti, D.; Johnson, J.; et al. Prognostic Significance of Tumor-Associated Macrophage Content in Head and Neck Squamous Cell Carcinoma: A Meta-Analysis. Front. Oncol. 2019, 9, 656. [Google Scholar] [CrossRef]
  100. Costa, N.L.; Valadares, M.C.; Souza, P.P.C.; Mendonça, E.F.; Oliveira, J.C.; Silva, T.A.; Batista, A.C. Tumor-Associated Macrophages and the Profile of Inflammatory Cytokines in Oral Squamous Cell Carcinoma. Oral Oncol. 2013, 49, 216–223. [Google Scholar] [CrossRef]
  101. JiaWei, Z.; ChunXia, D.; CunDong, L.; Yang, L.; JianKun, Y.; HaiFeng, D.; Cheng, Y.; ZhiPeng, H.; HongYi, W.; DeYing, L.; et al. M2 Subtype Tumor Associated Macrophages (M2-TAMs) Infiltration Predicts Poor Response Rate of Immune Checkpoint Inhibitors Treatment for Prostate Cancer. Ann. Med. 2021, 53, 730–740. [Google Scholar] [CrossRef]
  102. Economopoulou, P.; Kotsantis, I.; Psyrri, A. Tumor Microenvironment and Immunotherapy Response in Head and Neck Cancer. Cancers 2020, 12, 3377. [Google Scholar] [CrossRef] [PubMed]
  103. Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.-M.; Ries, C.H.; Rüttinger, D. Colony-Stimulating Factor 1 Receptor (CSF1R) Inhibitors in Cancer Therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef]
  104. Porcheri, C.; Mitsiadis, T.A. New Scenarios in Pharmacological Treatments of Head and Neck Squamous Cell Carcinomas. Cancers 2021, 13, 5515. [Google Scholar] [CrossRef] [PubMed]
  105. Method of the Year 2017: Organoids. Nat. Methods 2018, 15, 1. [CrossRef] [Green Version]
  106. Huang, M.; Lou, Y.; Pellissier, J.; Burke, T.; Liu, F.X.; Xu, R.; Velcheti, V. Cost Effectiveness of Pembrolizumab vs. Standard-of-Care Chemotherapy as First-Line Treatment for Metastatic NSCLC That Expresses High Levels of PD-L1 in the United States. PharmacoEconomics 2017, 35, 831–844. [Google Scholar] [CrossRef] [PubMed]
  107. Driehuis, E.; Kolders, S.; Spelier, S.; Lõhmussaar, K.; Willems, S.M.; Devriese, L.A.; de Bree, R.; de Ruiter, E.J.; Korving, J.; Begthel, H.; et al. Oral Mucosal Organoids as a Potential Platform for Personalized Cancer Therapy. Cancer Discov. 2019, 9, 852–871. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biomarkers and strategies to target oncogenic programs in HNSCC. (A) DNA methylation transferases (DNMT) promote DNA methylation and suppress the expression of immune programs. This could be reactivated through utilization of DNMT inhibitors (DNMTi). DNMTi when combined with anti-PD1 therapy could elicit better response in patients and improve survival. (B) Trametinib (MEK inhibitor) resistance and FAT1 mutation results in increased YAP1 mediated transcriptional programs. This could result in aggressive disease state in HNSCC. (C) Strategies utilizing single cell technologies and genomics can be utilized to address tumor heterogeneity and also identify novel transcriptional programs or biomarkers. This could help in development of novel therapeutic opportunities. The new drugs can be screened in patient-derived organoids and xenografts for their efficacy and ability to control tumor growth in HNSCC.
Figure 1. Biomarkers and strategies to target oncogenic programs in HNSCC. (A) DNA methylation transferases (DNMT) promote DNA methylation and suppress the expression of immune programs. This could be reactivated through utilization of DNMT inhibitors (DNMTi). DNMTi when combined with anti-PD1 therapy could elicit better response in patients and improve survival. (B) Trametinib (MEK inhibitor) resistance and FAT1 mutation results in increased YAP1 mediated transcriptional programs. This could result in aggressive disease state in HNSCC. (C) Strategies utilizing single cell technologies and genomics can be utilized to address tumor heterogeneity and also identify novel transcriptional programs or biomarkers. This could help in development of novel therapeutic opportunities. The new drugs can be screened in patient-derived organoids and xenografts for their efficacy and ability to control tumor growth in HNSCC.
Curroncol 29 00334 g001
Table 1. PD-L1 biomarker data in HNSCC.
Table 1. PD-L1 biomarker data in HNSCC.
StudiesTreatmentPD-L1 AssayPositivity
Cut-Off
OutcomesReferences
KEYNOTE-012
(NCT01848834)
PembrolizumabPD-L1 IHC
22C3 PharmDx
TPS ≥ 1
CPS ≥ 1
ORR (p = 0.023)
PD-L1+ (21%)
PD-L1− (6%)
OS (p = 0.008)
PD-L1+ (10 months)
PD-L1− (5 months)
[19,20]
KEYNOTE-055
(NCT02255097)
PembrolizumabPD-L1 IHC
22C3 PharmDx
CPS ≥ 1
CPS ≥ 50
ORR (CPS ≥ 1)
PD-L1+ (18%)
PD-L1− (12%)
ORR (CPS ≥ 50)
PD-L1+ (27%)
PD-L1− (13%)
[21,22]
KEYNOTE-040
(NCT02252042)
Pembrolizumab vs.
Investigator’s Choice (IC)
PD-L1 IHC
22C3 PharmDx
TPS ≥ 50
CPS ≥ 1
OS
Pembro vs. IC (TPS ≥ 50)
HR: 0.53, p = 0.0014
Pembro vs. IC (CPS ≥ 1)
HR: 0.74, p = 0.0049
[11,23,24,25]
CHECKMATE-141
(NCT02105636)
Nivolumab
vs.
Investigator’s Choice (IC)
PD-L1 IHC
28–8 PharmDx
TC ≥ 1OS
7.7 vs. 3.3 months,
HR: 0.56
12-month OS
39.2% vs. 15.4%
24-month OS
20.4% vs. 3.8%
ORR
20.0% vs. 11.5%
[26]
KEYNOTE-048
(NCT02358031)
Pembrolizumab
vs.
Pembrolizumab + chemotherapy
vs.
Cetuximab + chemotherapy
(Extreme)
PD-L1 IHC
22C3 PharmDx
CPS ≥ 1
CPS ≥ 20
OS
Pembro vs. Extreme
(CPS ≥ 20)
14.9 vs. 10.7 months
HR: 0.61, p = 0.0007
Pembro vs. Extreme
(CPS ≥ 1)
12.3 vs. 10.3 months
HR: 0.78, p = 0.0086
Pembro+ vs. Extreme
(CPS ≥ 20)
14.7 vs. 11.0 months
HR: 0.60, p = 0.0004
Pembro+ vs. Extreme
(CPS ≥ 1)
13.6 vs. 10.4 months
HR: 0.65, p < 0.0001
[27,28,29]
HAWKDurvalumabVENTANA
PD-L1 SP263
TC ≥ 25%ORR
Total: 16.2%
HPV+: 29.4%
HPV−: 10.9%
OS
mOS: 7.1 months
12-month OS: 33.6%
[30]
CONDORDurvalumab
vs.
tremelimumab
vs.
durvalumab + tremelimumab
VENTANA
PD-L1 SP263
TC ≥ 25%Durvalumab
ORR: 9.2%
Tremelimumab
ORR: 1.6%
Durvalumab + Tremelimumab
ORR: 7.8%
[31]
EAGLEDurvalumab
vs.
Durvalumab + tremelimumab
vs.
Investigator’s Choice (IC)
VENTANA
PD-L1 SP263
TC ≥ 25%OS
Durvalumab
PD-L1+ (9.8 months)
PD-L1− (7.6 months)
Durvalumab + Tremelimumab
PD-L1+ (4.8 months)
PD-L1− (7.8 months)
IC
PD-L1+ (9.0 months)
PD-L1− (8.0 months)
[32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, J.C.; Krishnakumar, H.N.; Saladi, S.V. Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma. Curr. Oncol. 2022, 29, 4185-4198. https://doi.org/10.3390/curroncol29060334

AMA Style

Park JC, Krishnakumar HN, Saladi SV. Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma. Current Oncology. 2022; 29(6):4185-4198. https://doi.org/10.3390/curroncol29060334

Chicago/Turabian Style

Park, Jong Chul, Hari N. Krishnakumar, and Srinivas Vinod Saladi. 2022. "Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma" Current Oncology 29, no. 6: 4185-4198. https://doi.org/10.3390/curroncol29060334

Article Metrics

Back to TopTop