Next Article in Journal
Multivariate Statistical Approach for Nephrines in Women with Obesity
Next Article in Special Issue
The Glycolytic Pathway as a Target for Novel Onco-Immunology Therapies in Pancreatic Cancer
Previous Article in Journal
GC-FID-MS Based Metabolomics to Access Plum Brandy Quality
Previous Article in Special Issue
The Microbiome and Its Implications in Cancer Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current Landscape of Immune Checkpoint Blockade in Metastatic Lung Squamous Cell Carcinoma

1
Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2
State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
*
Authors to whom correspondence should be addressed.
Submission received: 24 January 2021 / Revised: 21 February 2021 / Accepted: 2 March 2021 / Published: 5 March 2021
(This article belongs to the Special Issue The Future of the Cancer Treatment: The Immunotherapy Next Generation)

Abstract

:
In addition to surgery, chemotherapy, radiotherapy, and targeted therapy, immunotherapy has emerged as a standard pillar of cancer treatment. Immune checkpoint inhibitors (ICIs) such as targeting programmed death-1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have been integrated into standard-of-care regimens for patients with advanced lung squamous cell carcinoma (LUSC), who were previously limited by the lack of treatment options. Atezolizumab, durvalumab, nivolumab, and pembrolizumab are all currently used as part of standard-of-care treatment for different stages of lung cancer. Recent successes and failures of immune checkpoint blockade-based combination therapies have provided significant insights into implementing combination strategies in LUSC. Therefore, there is an urgent need to correctly select patients who are more likely to respond to immunotherapy and understand the mechanisms of primary or acquired resistance. In this review, we aim at summarizing the emerging clinical data on the promise and challenge of ICIs, discussing the unmet need of potential biomarkers for predicting response or resistance to immunotherapy, and providing an overview of the current immune landscape and future directions in advanced LUSC.

1. Introduction

Lung cancer is the leading cause of cancer-related mortality all over the world [1,2]. Lung squamous cell carcinoma (LUSC), a subtype of non-small cell lung cancer (NSCLC), comprises approximately 30% of NSCLC and is attributable to smoking. Development of squamous cell carcinoma (SCC) is related to genomic perturbations, gene mutations, chromosomal instabilities, and/or altered expression of key molecules involved in different stages of squamous cell lineage commitment and/or terminal differentiation.
Patients with driver mutation positive lung adenocarcinoma (LUAD) can benefit from a targeted therapy such as tyrosine kinase inhibitors (TKIs), while few druggable targets have been identified in LUSC [3,4]. Standard platinum-based doublet chemotherapy has always represented the first-line therapy for advanced/metastatic LUSC [5,6].
The FLEX trial showed that overall survival (OS) was prolonged in a chemotherapy plus cetuximab group (11.3 months) versus a chemotherapy alone group (10.1 months) in patients with advanced NSCLC across all histological subtypes (adenocarcinoma, squamous cell carcinoma, etc.) [7]. The SQUIRE trial demonstrated that the addition of necitumumab, a second-generation EGFR antibody, to platinum-based doublet chemotherapy improved survival (mOS, 11.5 months versus 9.9 months) in patients with previously untreated stage IV squamous NSCLC [8]. However, the survival benefit from the combination of chemotherapy and targeted therapy was limited for advanced LUSC patients.
Lee J.S. indicated that anti-programmed death-1/programmed death ligand 1 (anti-PD-1/PD-L1) therapy can provide significant survival benefits and a high objective response rate (ORR) for LUSC patients [9]. The emergence of immune checkpoint inhibitors (ICIs) has revolutionized the standard care of patients with LUSC, previously limited by the lack of treatment selections [10]. Motivated by this progress, a large number of immunotherapy-based studies have currently been implemented worldwide, alone or in combination with other systemic therapies.
In this review, we summarize PD-1/PD-L1 inhibitors in clinical practice (Table 1), recent clinical trials on ICIs in LUSC, and discuss the need for biomarkers for predicting potential response or resistance to immunotherapies.

2. Immune Checkpoint Inhibitors (ICIs) in Advanced Lung Squamous Cell Carcinoma (LUSC) after First-Line Therapy Failure

Immunotherapy is widely used in the care of patients with NSCLC, and multiple standard-of-care (SOC) regimens are approved in metastatic LUSC (Table 2). Studies such as KeyNote-010 [11], CheckMate 017 [12,13,14] /078 [15], POPLAR [16], and OAK [17,18] have proven that ICIs can improve the prognosis of patients with advanced NSCLC, bringing about a new alternative for second-line treatment.
Studies have evaluated single agent PD-1/PD-L1 blockade in previously treated, advanced NSCLC and have demonstrated improved efficacy over standard chemotherapy with docetaxel. Nivolumab, pembrolizumab, and atezolizumab were approved based on the results of CheckMate 017 [12], KeyNote-010 [11], and OAK [17,18], respectively. Compared with that in chemotherapy, the hazard ratio (HR) for the OS of ICIs ranged from 0.59 to 0.73. Importantly, the significant improvement in the OS came with notable reductions in toxicity. The incidence of severe or life-threatening adverse events (grade 3 or higher) ranged from 7% to 15% with ICIs versus 35% to 55% as compared with docetaxel.

3. ICIs in Advanced LUSC in First-Line Treatment

Treatment with PD-1/PD-L1 blockade has now become the front-line therapy in patients with lung cancer (Table 3). In the KeyNote-024 study [19], patients with PD-L1 high expression, tumor proportion score (TPS) > 50%, and without targetable alterations in epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) were randomized to receive either pembrolizumab or platinum-based doublet chemotherapy. The median progression-free survival (PFS) was significantly longer in the pembrolizumab group than that of standard platinum-based doublet chemotherapy (10.3 vs. 6 months, HR 0.50, 95% CI 0.37–0.68, p < 001). From longer term follow-up data, the median OS for the pembrolizumab group was 30 months as compared with 14.2 months in the chemotherapy group (HR 0.49, 95% CI 0.34–0.69).
For those patients who may not tolerate chemotherapy and have PD-L1 expression (TPS > 1%), single-agent pembrolizumab may be taken into consideration based on KeyNote-042 [20], although the benefit of this approach was largely driven by patients with high expression of PD-L1.
In KeyNote-407 study [21], patients were randomized to a chemotherapy backbone of carboplatin and investigator’s choice of either paclitaxel or nanoparticle-bound paclitaxel plus pembrolizumab or placebo. The median OS was 15.9 months with pembrolizumab plus chemotherapy versus 11.3 months with placebo plus chemotherapy (HR for death, 0.64, 95% CI 0.49–0.85, p < 001). This combination is applicable to patients with either squamous or non-squamous cell carcinoma regardless of PD-L1 expression level.
IMpower 131 study [22] revealed that atezolizumab in combination with carboplatin and nab-paclitaxel (A + CnP) as first-line treatment, significantly improved PFS as compared with chemotherapy alone in patients with stage IV squamous NSCLC, but the OS was similar among A + CP (atezolizumab+ carboplatin+ paclitaxel), A + CnP, and CnP (carboplatin + nab-paclitaxel) arms.
Front-line nivolumab plus ipilimumab was compared with chemotherapy in the CheckMate 227 trial [23,24,25] and the results indicated that chemotherapy free might be an option for patients who showed intolerance to chemotherapy and were not optimal candidates for single-agent anti-PD-1/PD-L1 therapy. This study enrolled patients with advanced, metastatic NSCLC stratified by PD-L1 expression ≥1% versus <1% and randomized 1:1:1 to nivolumab plus ipilimumab, nivolumab alone, or platinum doublet chemotherapy. An earlier report of the first co-primary endpoint of the study highlighted high tumor mutational burden (TMB) (≥10 mutations/Mb) as a biomarker predicting longer PFS with combination immunotherapy; however, this result did not persist in the OS analysis. In the second co-primary endpoint, across PD-L1 expression levels, the median OS was longer in the immunotherapy group than that in the chemotherapy group. Specifically, in patients with PD-L1 expression (TPS ≥ 1%), the median OS was 17.1 months (95% CI, 15.0–20.1 months) with nivolumab plus ipilimumab versus 14.9 months (95% CI, 12.7–16.7 months) with chemotherapy, whereas in the PD-L1 < 1% group, the median OS was 17.2 months (95% CI, 12.8–22.0 months) versus 12.2 months (95% CI, 9.2–14.3 months), respectively.

4. Immune Checkpoint Blockade-Based Combination Strategies

PD-L1 and programmed death ligand 2 (PD-L2) are ligands of PD-1 and the interaction of PD-L1 or PD-L2 with PD-1 results in T cell suppression. The affinity of PD-L2/PD-1 is 2~6-fold higher than the interaction of PD-L1/PD-1 [26]. Additionally, the binding of PD-L1 to the cluster of differentiation 80 (CD80) on activated T cells can inhibit T cell activity. Conversely, PD-L2 can interact with repulsive guidance molecule B (RGMB) receptor, a co-receptor for bone morphogenetic protein (BMP), resulting in the activation of T cells [27].
PD-1 antibodies can block PD-1/PD-L1 and PD-1/PD-L2 pathways but leave the PD-L1/CD80 axis unaffected, leading to the inhibition of T cell activation. In addition, anti-PD-1 indirectly enhances PD-L2/RGMB interaction, resulting in the improvement of T cell-mediated anti-tumor immune response. Unlike the PD-1 antibodies, the PD-L1 antibodies also block the PD-1/PD-L1 and PD-L1/CD80 axes but leave the PD-L2/PD-1 axis unaffected, leading to tumor immune escape [28]. Such evolving knowledge of the biochemical and signaling effects will shed light on the molecular mechanisms of PD-1 checkpoint blockade-responsive patients and guide the design of combination therapies to modulate PD-1 and its downstream targets.
Immune checkpoint blockers activate effector T cells, which in turn facilitate the normalization of tumor blood vessels. The tumor vascular normalization promotes effector T cells infiltration into tumors and improves their function, leading to the enhancement of antitumor immunity [29]. This feedback loop between immune reprogramming and tumor vascular normalization reinforces itself, ultimately promoting immune-mediated tumor eradication [30]. The immune reprogramming-tumor vasculature crosstalk provides us with a rationale for combination approaches using immunotherapy and antiangiogenic therapy [31].
Clinical trials combining PD-1/PD-L1 blockade with cytotoxic therapy or with other ICI strategies have shown higher response rates at the risk of higher toxicity. According to the CheckMate 227 study, nivolumab plus ipilimumab was an option for first-line treatment of advanced NSCLC without driver mutations in patients with PD-L1 TPS ≥ 1%. However, there is no head-to-head comparison of anti-PD-1 plus chemotherapy versus anti-PD-L1 plus chemotherapy for advanced LUSC patients. A meta-analysis using data from the KeyNote-407 and IMpower 131 studies revealed that pembrolizumab brought significantly better OS (HR 0.67, p = 0.02) and numerically superior PFS (HR 0.79, p = 0.10) as compared with atezolizumab in LUSC patients [32]. Therefore, studies have been implemented to explore the optimal choice of immunotherapy (anti-PD-1, anti-PD-L1, or anti-CTLA-4) for advanced LUSC patients in combination with chemotherapy [33,34,35].

5. Biomarkers Predicting the Efficacy of Immunotherapy

Only a minority of patients have achieved sustained benefits from ICI; reliable biomarkers that can accurately identify these patients before or early during treatment have thus far remained elusive. Biomarkers such as PD-L1 (Table 4), TMB (Table 5), or tumor inflammation, offer insight into which patients may benefit most from the emerging agents and combination strategies. PD-L1 has several major shortcomings as a predictive biomarker of durable benefit while TMB is continuing to be evaluated clinically.
The current practice in the USA for front-line therapy of driver mutation-negative advanced NSCLC is to treat patients with PD-L1 ≤ 49% (percentage of PD-L1 positive tumor cells by immunohistochemistry) with concurrent chemotherapy plus pembrolizumab, and those with PD-L1 ≥ 50% with pembrolizumab alone or with concurrent chemotherapy.
Studies have shown that ICIs improve overall survival as compared with chemotherapy in first-line therapy for patients with PD-L1 expression ≥ 50%. A combination of ICIs with chemotherapy improves survival in patients with both squamous and non-squamous NSCLC, regardless of the expression level of PD-L1. However, a subset of patients with PD-L1 TPS < 1% can benefit from immunotherapy alone [12], suggesting that PD-L1 is an imperfect predictive biomarker. PD-L1 immunohistochemistry (IHC) staining alone is not sufficiently accurate to identify potential responders to PD-1/PD-L1 blockade-based immunotherapy in NSCLC [36,37]. In addition, heterogeneity within tumors, biopsies from primary versus metastatic lesions, different detection antibodies and cut-offs, staining procedures, and immune escape to PD-L1/PD-1 blockade are key factors that likely relate to the predictive value of PD-L1 [38,39,40]. One approach to resolve the limitation of spatial resolution involves positron emission tomography (PET)-based PD-L1 imaging with zirconium-89-labeled atezolizumab [41,42]. The technique, a noninvasive imaging of tumor PD-L1 expression in vivo, may enable patient selection from anti-PD-1/PD-L1 therapy and monitor PD-L1 expression during therapy [41]. Bensch F. et al. presented the initial results from a first-in-human study to assess the feasibility of imaging with zirconium-89-labeled atezolizumab and tested its potential to predict a clinical response to PD-L1 blockade in NSCLC [42].
CheckMate 227 (nivolumab plus ipilimumab versus platinum-doublet chemotherapy as first-line therapy for advanced NSCLC) displayed that TMB correlated with the clinical response to combination therapy with nivolumab and ipilimumab as a first-line regimen for NSCLC [23]. The CheckMate 227 trial was the first study that evaluated TMB as a biomarker in NSCLC patients who received immunotherapy. A significant benefit such as prolonged PFS was not observed in the overall population who received the combination therapy as compared with chemotherapy alone. Patients with TMB ≥ 10 mutations/Mb who received the combination treatment showed significantly better PFS as compared with those who received chemotherapy alone, irrespective of the expression of PD-L1. TMB might be a predictive biomarker for NSCLC patients who would benefit from ICB-based therapy [43].
In the future, tumor biopsy tissue collected prior to treatment, on-treatment, and disease progression will likely enable greater sophistication in treatment selection and achieve immune eradication of tumors.

6. Strategies Overcoming Immunotherapy Resistance

The biology underlying resistance is driven by the interplay between the immune system and tumor microenvironment (TME) and is influenced by multiple factors. Patients who have disease progression after receiving at least 6 weeks of exposure to PD-1/PD-L1 checkpoint inhibitors, but with PD/SD for ≤6 months can be considered to have primary resistant disease. Patients with SD for ≥6 months, who then had overall disease progression, independent of the target lesion measurements (overall tumor regression of <30% or tumor growth <20%), would be considered to have secondary resistance [44].
ICIs-targeted regimens can fail patients because of primary resistance, when a cancer does not respond to immunotherapy, or acquired resistance, i.e., when a cancer initially responds but then progresses [44,45]. Constitutive mechanisms of resistance to ICB therapies include innate PD-1 resistance signature (IPRES), oncogene activation (Wnt/β-catenin), loss of oncosuppressor genes (PTEN), downregulation of the type I IFN receptor IFNAR1 on cytotoxic T lymphocytes (CTLs), and poor infiltration by CD8+ T cells. Acquired resistance to ICIs includes disruption/downregulation of antigen presentation machinery, loss of IFN-γ sensitivity, neoantigen depletion, tumor-mediated immune suppression or immune exclusion, and additional inhibitory checkpoints [45].
Among tumor types, there appears to be an inverse association between overall response rate and the frequency of acquired resistance among responders to PD-1 blockade [46]. Strategies to combat resistance to immunotherapy involve enhancing antigenicity, modulation of TME, increasing immune cell activity, and overcoming resistance mediated by other upregulated immune checkpoints.
Mechanism-based strategies to overcome resistance to immune checkpoint targeted therapies (ICT) are urgently needed. Ishizuka J.J. et al. showed that loss of RNA-editing enzyme ADAR1 overcomes resistance to PD-1 checkpoint blockade caused by inactivation of antigen presentation in tumor cells [47]. Cortez M.A. et al. found that BMP7 impairs proinflammatory responses in the tumor microenvironment by inhibiting mitogen-activated protein kinase 14 (MAPK14) expression in macrophages and CD4+ T cells. Knockdown of BMP7 in combination with anti-PD1 activates CD4+ and CD8+ T cells in tumors, decreases M2 macrophages, and resensitizes resistant tumors to immunotherapies [48]. Emerging evidence demonstrates that targeting epigenetic elements that promote tumor progression and inhibit immune cell activity can enhance antitumor immunity by reshaping the TME. Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 (PRC2), inhibition can increase T regulatory cell trafficking, impair T regulatory cell capacity, increase antigen presentation, and increase antitumor immunity, making it a promising target for overcoming ICB resistance of certain cancers [49].
The search for better prognostic and predictive biomarkers is ongoing and will be essential for improving patient selection for the growing list of therapeutic options [50].

7. Conclusions

In-depth tumor analysis including whole-genome sequencing, single cell RNA-sequencing, multidimensional flow cytometry, or epigenetics might be implemented in the future in order to find individualized treatment strategies.
Consideration of common properties across SCCs as they relate to epigenetics, genomics, genetics, and transcriptomes may serve as a foundation for individual and combinatorial therapeutics, as well as understanding the mechanistic basis of treatment resistance.
The emergence of PD-1/PD-L1/CTLA-4-targeted therapy has revolutionized the standard care of LUSC patients, generating durable responses and prolonged overall survival. However, not all patients benefit from immunotherapy. Primary or acquired resistance prevents patients from experiencing tumor shrinkage or extended survival. Predictive biomarkers such as PD-L1, TMB, or tumor inflammation, deserve further study.
An improved understanding of the biology and molecular subtypes of non-small cell lung cancer has resulted in the development of new biomarker-directed therapies and has led to improvements in overall survival of patients with advanced or metastatic disease. Since the introduction of PD-1/PD-L1 blockers in 2015, patients without molecular therapeutic targets now receive treatment with one of the immune checkpoint therapies in the first-line setting, especially for squamous NSCLC patients. Compared with previous studies, we have focused on the landscape of ICB in metastatic lung squamous cell carcinoma [43,51,52].
In conclusion, immunotherapy has changed the SOC for NSCLC, and has paved the way for a new treatment paradigm. Despite these advances, monumental efforts are still needed to significantly improve the outcome of patients with advanced/metastatic LUSC.

Funding

This research was funded by National Science Foundation of China, grant number 81972707 and Shanghai Anticancer Association EYAS PROJECT, grant number SACA-CY19C19.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the members of the Department of Oncology within Ruijin Hospital, Shanghai Jiao Tong University School of Medicine for helpful discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA A Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2021. [Google Scholar] [CrossRef] [PubMed]
  3. Drilon, A.; Rekhtman, N.; Ladanyi, M.; Paik, P. Squamous-cell carcinomas of the lung: Emerging biology, controversies, and the promise of targeted therapy. Lancet Oncol. 2012, 13, e418–e426. [Google Scholar] [CrossRef]
  4. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012, 489, 519–525. [CrossRef] [PubMed]
  5. Socinski, M.A.; Bondarenko, I.; Karaseva, N.A.; Makhson, A.M.; Vynnychenko, I.; Okamoto, I.; Hon, J.K.; Hirsh, V.; Bhar, P.; Zhang, H.; et al. Weekly nab-paclitaxel in combination with carboplatin versus solvent-based paclitaxel plus carboplatin as first-line therapy in patients with advanced non-small-cell lung cancer: Final results of a phase III trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 2055–2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lu, S.; Chen, Z.; Hu, C.; Zhang, J.; Chen, Y.; Song, Y.; Zhao, Q.; Fan, Y.; Wu, G.; Ma, Z.; et al. Nedaplatin Plus Docetaxel Versus Cisplatin Plus Docetaxel as First-Line Chemotherapy for Advanced Squamous Cell Carcinoma of the Lung—A Multicenter, Open-label, Randomized, Phase III Trial. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2018, 13, 1743–1749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Pirker, R.; Pereira, J.R.; Szczesna, A.; von Pawel, J.; Krzakowski, M.; Ramlau, R.; Vynnychenko, I.; Park, K.; Yu, C.T.; Ganul, V.; et al. Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): An open-label randomised phase III trial. Lancet 2009, 373, 1525–1531. [Google Scholar] [CrossRef]
  8. Thatcher, N.; Hirsch, F.R.; Luft, A.V.; Szczesna, A.; Ciuleanu, T.E.; Dediu, M.; Ramlau, R.; Galiulin, R.K.; Bálint, B.; Losonczy, G.; et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): An open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015, 16, 763–774. [Google Scholar] [CrossRef]
  9. Lee, J.S.; Ruppin, E. Multiomics Prediction of Response Rates to Therapies to Inhibit Programmed Cell Death 1 and Programmed Cell Death 1 Ligand 1. JAMA Oncol. 2019, 5, 1614–1618. [Google Scholar] [CrossRef] [PubMed]
  10. Zhang, X.C.; Wang, J.; Shao, G.G.; Wang, Q.; Qu, X.; Wang, B.; Moy, C.; Fan, Y.; Albertyn, Z.; Huang, X.; et al. Comprehensive genomic and immunological characterization of Chinese non-small cell lung cancer patients. Nat. Commun. 2019, 10, 1772. [Google Scholar] [CrossRef] [Green Version]
  11. Herbst, R.S.; Baas, P.; Kim, D.W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.Y.; Molina, J.; Kim, J.H.; Arvis, C.D.; Ahn, M.J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef]
  12. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Horn, L.; Spigel, D.R.; Vokes, E.E.; Holgado, E.; Ready, N.; Steins, M.; Poddubskaya, E.; Borghaei, H.; Felip, E.; Paz-Ares, L.; et al. Nivolumab Versus Docetaxel in Previously Treated Patients With Advanced Non-Small-Cell Lung Cancer: Two-Year Outcomes From Two Randomized, Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057). J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 3924–3933. [Google Scholar] [CrossRef] [PubMed]
  14. Vokes, E.E.; Ready, N.; Felip, E.; Horn, L.; Burgio, M.A.; Antonia, S.J.; Arén Frontera, O.; Gettinger, S.; Holgado, E.; Spigel, D.; et al. Nivolumab versus docetaxel in previously treated advanced non-small-cell lung cancer (CheckMate 017 and CheckMate 057): 3-year update and outcomes in patients with liver metastases. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 959–965. [Google Scholar] [CrossRef]
  15. Wu, Y.L.; Lu, S.; Cheng, Y.; Zhou, C.; Wang, J.; Mok, T.; Zhang, L.; Tu, H.Y.; Wu, L.; Feng, J.; et al. Nivolumab Versus Docetaxel in a Predominantly Chinese Patient Population With Previously Treated Advanced NSCLC: CheckMate 078 Randomized Phase III Clinical Trial. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2019, 14, 867–875. [Google Scholar] [CrossRef]
  16. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  17. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
  18. Gandara, D.R.; von Pawel, J.; Mazieres, J.; Sullivan, R.; Helland, Å.; Han, J.Y.; Ponce Aix, S.; Rittmeyer, A.; Barlesi, F.; Kubo, T.; et al. Atezolizumab Treatment beyond Progression in Advanced NSCLC: Results From the Randomized, Phase III OAK Study. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2018, 13, 1906–1918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
  20. Mok, T.S.K.; Wu, Y.L.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
  21. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef] [PubMed]
  22. Jotte, R.; Cappuzzo, F.; Vynnychenko, I.; Stroyakovskiy, D.; Rodríguez-Abreu, D.; Hussein, M.; Soo, R.; Conter, H.J.; Kozuki, T.; Huang, K.C.; et al. Atezolizumab in Combination With Carboplatin and Nab-Paclitaxel in Advanced Squamous NSCLC (IMpower131): Results From a Randomized Phase III Trial. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2020, 15, 1351–1360. [Google Scholar] [CrossRef]
  23. Hellmann, M.D.; Ciuleanu, T.E.; Pluzanski, A.; Lee, J.S.; Otterson, G.A.; Audigier-Valette, C.; Minenza, E.; Linardou, H.; Burgers, S.; Salman, P.; et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N. Engl. J. Med. 2018, 378, 2093–2104. [Google Scholar] [CrossRef] [PubMed]
  24. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef]
  25. Reck, M.; Schenker, M.; Lee, K.H.; Provencio, M.; Nishio, M.; Lesniewski-Kmak, K.; Sangha, R.; Ahmed, S.; Raimbourg, J.; Feeney, K.; et al. Nivolumab plus ipilimumab versus chemotherapy as first-line treatment in advanced non-small-cell lung cancer with high tumour mutational burden: Patient-reported outcomes results from the randomised, open-label, phase III CheckMate 227 trial. Eur. J. Cancer 2019, 116, 137–147. [Google Scholar] [CrossRef]
  26. Boussiotis, V.A. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N. Engl. J. Med. 2016, 375, 1767–1778. [Google Scholar] [CrossRef] [Green Version]
  27. Larsen, T.V.; Hussmann, D.; Nielsen, A.L. PD-L1 and PD-L2 expression correlated genes in non-small-cell lung cancer. Cancer Commun. 2019, 39, 30. [Google Scholar] [CrossRef] [Green Version]
  28. Duan, J.; Cui, L.; Zhao, X.; Bai, H.; Cai, S.; Wang, G.; Zhao, Z.; Zhao, J.; Chen, S.; Song, J.; et al. Use of Immunotherapy With Programmed Cell Death 1 vs Programmed Cell Death Ligand 1 Inhibitors in Patients With Cancer: A Systematic Review and Meta-analysis. JAMA Oncol. 2020, 6, 375–384. [Google Scholar] [CrossRef]
  29. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Huang, Y.; Kim, B.Y.S.; Chan, C.K.; Hahn, S.M.; Weissman, I.L.; Jiang, W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 195–203. [Google Scholar] [CrossRef]
  31. Liu, Z.; Wang, Y.; Huang, Y.; Kim, B.Y.S.; Shan, H.; Wu, D.; Jiang, W. Tumor Vasculatures: A New Target for Cancer Immunotherapy. Trends Pharmacol. Sci. 2019, 40, 613–623. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, Y.; Zhou, H.; Zhang, L. Which is the optimal immunotherapy for advanced squamous non-small-cell lung cancer in combination with chemotherapy: Anti-PD-1 or anti-PD-L1? J. Immunother. Cancer 2018, 6, 135. [Google Scholar] [CrossRef]
  33. Zappasodi, R.; Merghoub, T.; Wolchok, J.D. Emerging Concepts for Immune Checkpoint Blockade-Based Combination Therapies. Cancer Cell 2018, 33, 581–598. [Google Scholar] [CrossRef] [Green Version]
  34. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  35. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
  36. Phillips, T.; Simmons, P.; Inzunza, H.D.; Cogswell, J.; Novotny, J., Jr.; Taylor, C.; Zhang, X. Development of an automated PD-L1 immunohistochemistry (IHC) assay for non-small cell lung cancer. Appl. Immunohistochem. Mol. Morphol. Aimm 2015, 23, 541–549. [Google Scholar] [CrossRef] [Green Version]
  37. McLaughlin, J.; Han, G.; Schalper, K.A.; Carvajal-Hausdorf, D.; Pelekanou, V.; Rehman, J.; Velcheti, V.; Herbst, R.; LoRusso, P.; Rimm, D.L. Quantitative Assessment of the Heterogeneity of PD-L1 Expression in Non-Small-Cell Lung Cancer. JAMA Oncol. 2016, 2, 46–54. [Google Scholar] [CrossRef] [PubMed]
  38. Kim, J.M.; Chen, D.S. Immune escape to PD-L1/PD-1 blockade: Seven steps to success (or failure). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1492–1504. [Google Scholar] [CrossRef] [PubMed]
  39. Lanitis, E.; Dangaj, D.; Irving, M.; Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, xii18–xii32. [Google Scholar] [CrossRef] [PubMed]
  40. Hong, L.; Negrao, M.V.; Dibaj, S.S.; Chen, R.; Reuben, A.; Bohac, J.M.; Liu, X.; Skoulidis, F.; Gay, C.M.; Cascone, T.; et al. Programmed Death-Ligand 1 Heterogeneity and Its Impact on Benefit From Immune Checkpoint Inhibitors in NSCLC. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2020, 15, 1449–1459. [Google Scholar] [CrossRef]
  41. Heskamp, S.; Hobo, W.; Molkenboer-Kuenen, J.D.; Olive, D.; Oyen, W.J.; Dolstra, H.; Boerman, O.C. Noninvasive Imaging of Tumor PD-L1 Expression Using Radiolabeled Anti-PD-L1 Antibodies. Cancer Res. 2015, 75, 2928–2936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Bensch, F.; van der Veen, E.L.; Lub-de Hooge, M.N.; Jorritsma-Smit, A.; Boellaard, R.; Kok, I.C.; Oosting, S.F.; Schröder, C.P.; Hiltermann, T.J.N.; van der Wekken, A.J.; et al. (89)Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat. Med. 2018, 24, 1852–1858. [Google Scholar] [CrossRef] [PubMed]
  43. Doroshow, D.B.; Sanmamed, M.F.; Hastings, K.; Politi, K.; Rimm, D.L.; Chen, L.; Melero, I.; Schalper, K.A.; Herbst, R.S. Immunotherapy in Non-Small Cell Lung Cancer: Facts and Hopes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4592–4602. [Google Scholar] [CrossRef] [Green Version]
  44. Kluger, H.M.; Tawbi, H.A.; Ascierto, M.L.; Bowden, M.; Callahan, M.K.; Cha, E.; Chen, H.X.; Drake, C.G.; Feltquate, D.M.; Ferris, R.L.; et al. Defining tumor resistance to PD-1 pathway blockade: Recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [Green Version]
  45. Bellone, M.; Elia, A.R. Constitutive and acquired mechanisms of resistance to immune checkpoint blockade in human cancer. Cytokine Growth Factor Rev. 2017, 36, 17–24. [Google Scholar] [CrossRef] [PubMed]
  46. Schoenfeld, A.J.; Hellmann, M.D. Acquired Resistance to Immune Checkpoint Inhibitors. Cancer Cell 2020, 37, 443–455. [Google Scholar] [CrossRef]
  47. Ishizuka, J.J.; Manguso, R.T.; Cheruiyot, C.K.; Bi, K.; Panda, A.; Iracheta-Vellve, A.; Miller, B.C.; Du, P.P.; Yates, K.B.; Dubrot, J.; et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 2019, 565, 43–48. [Google Scholar] [CrossRef] [PubMed]
  48. Cortez, M.A.; Masrorpour, F.; Ivan, C.; Zhang, J.; Younes, A.I.; Lu, Y.; Estecio, M.R.; Barsoumian, H.B.; Menon, H.; Caetano, M.D.S.; et al. Bone morphogenetic protein 7 promotes resistance to immunotherapy. Nat. Commun. 2020, 11, 4840. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, H.J.; Cantor, H.; Cosmopoulos, K. Overcoming Immune Checkpoint Blockade Resistance via EZH2 Inhibition. Trends Immunol. 2020, 41, 948–963. [Google Scholar] [CrossRef]
  50. Havel, J.J.; Chowell, D.; Chan, T.A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 2019, 19, 133–150. [Google Scholar] [CrossRef] [PubMed]
  51. Arbour, K.C.; Riely, G.J. Systemic Therapy for Locally Advanced and Metastatic Non-Small Cell Lung Cancer: A Review. JAMA 2019, 322, 764–774. [Google Scholar] [CrossRef] [PubMed]
  52. Carlisle, J.W.; Steuer, C.E.; Owonikoko, T.K.; Saba, N.F. An update on the immune landscape in lung and head and neck cancers. Ca A Cancer J. Clin. 2020, 70, 505–517. [Google Scholar] [CrossRef] [PubMed]
Table 1. Programmed death-1/programmed death ligand 1 (PD-1/PD-L1) inhibitors were used in clinical practice for lung squamous cell carcinoma (LUSC) patients.
Table 1. Programmed death-1/programmed death ligand 1 (PD-1/PD-L1) inhibitors were used in clinical practice for lung squamous cell carcinoma (LUSC) patients.
DrugTargetFDA IndicationEMA IndicationNMPA Indication
Pembrolizumab (Keytruda)PD-1Pembrolizumab plus standard chemotherapy (carboplatin and either paclitaxel or nab-paclitaxel) for first-line treatment in patients with metastatic LUSC;
Approved for treatment of advanced or metastatic NSCLC patients with disease progression after platinum-based chemotherapy and PD-L1 positive
Approved for first-line treatment of metastatic NSCLC patients with PD-L1 TPS ≥ 50%, without a sensitizing EGFR mutation or ALK translocation;
Approved for treatment of locally advanced or metastatic NSCLC patients with PD-L1 TPS ≥ 1%, and disease progression during/after first-line chemotherapy
Approved for first-line treatment of locally advanced or metastatic NSCLC patients with PD-L1 TPS ≥ 1%, without a sensitizing EGFR mutation or ALK translocation;
Pembrolizumab plus standard chemotherapy (carboplatin and either paclitaxel or nab-paclitaxel) for first-line treatment in patients with metastatic LUSC
Nivolumab (Opdivo)PD-1Approved for treatment of patients with previously treated advanced NSCLC whose disease progression during/after platinum-based doublet chemotherapyApproved for treatment of patients with locally advanced or metastatic NSCLC whose disease progression during/after chemotherapyApproved for second-line or late-line treatment of patients with locally advanced or metastatic NSCLC without a sensitizing EGFR mutation or ALK translocation, and disease progression during/after platinum-based doublet chemotherapy
Tislelizumab
(TIZLEIO)
PD-1NANATislelizumab plus standard chemotherapy (carboplatin and paclitaxel) for first-line treatment of locally advanced or metastatic LUSC patients
Atezolizumab (Tecentriq)PD-L1Approved for treatment of patients with metastatic NSCLC whose disease progression during or after platinum-based chemotherapyApproved for treatment of patients with locally advanced or metastatic NSCLC whose disease progression during/after chemotherapyNA
Abbreviations: PD-1, programmed death-1; PD-L1, programmed death ligand 1; LUSC, lung squamous cell carcinoma; FDA, Food and Drug Administration; EMA, European Medicines Agency; NMPA, National Medical Products Administration; NSCLC, non-small cell lung cancer; TPS, tumor proportion score; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; NA, not available.
Table 2. The second-line clinical trials of anti-PD-1/PD-L1 in metastatic LUSC.
Table 2. The second-line clinical trials of anti-PD-1/PD-L1 in metastatic LUSC.
TrialsCheckMate 017CheckMate 078KeyNote 010POPLAROAK
HistologyLUSCNSCLCNSCLC (TPS ≥ 1%)NSCLCNSCLC
Treatment
Comparison
Nivolumab
Docetaxel
Divolumab
Docetaxel
Pembrolizumab DocetaxelAtezolizumab
Docetaxel
Atezolizumab Docetaxel
ORR20% vs. 8%16.6% vs. 4.2%19% vs. 10%NA14% vs. 13%
mOS
Squamous
Non-squamous
9.2 m vs. 6.0 m
/
/
12.0 m vs. 9.6 m
12.3 m vs. 7.9 m
11.9 m vs. 10.2 m
12.7 m vs. 10.4 m
/
/
12.6 m vs. 9.7 m
10.1 m vs. 8.6 m
15.5 m vs. 10.9 m
13.8 m vs. 9.6 m
8.9 m vs. 7.7 m
15.6 m vs. 11.2 m
OS HR0.59 (0.44–0.79)0.68 (0.52–0.90)0.71 (0.58–0.88)0.73 (0.53–0.99)0.73 (0.62–0.87)
Abbreviations: PD-1, programmed death-1; PD-L1, programmed death ligand 1; LUSC, lung squamous cell carcinoma; NSCLC, non-small cell lung cancer; TPS, tumor proportion score; ORR, objective response rate; mOS, median overall survival; HR, hazard ratio.
Table 3. The first-line clinical trials of immune checkpoint blockers in advanced LUSC.
Table 3. The first-line clinical trials of immune checkpoint blockers in advanced LUSC.
TrialsORIENT-12RATIONALE 307KeyNote-407IMpower 131
HistologyLUSCLUSCLUSCLUSC
GroupExperimental GroupControl GroupExperimental Group AExperimental Group BControl GroupExperimental GroupControl GroupExperimental GroupControl Group
N179178120119121278281343340
TreatmentSintilimab
gemcitabine
cisplatin/
carboplatin
Placebo
gemcitabine cisplatin/
carboplatin
Tislelizumab carboplatin paclitaxelTislelizumab carboplatin nab-paclitaxelCarboplatin paclitaxelPembrolizumab carboplatin paclitaxel/
nab-paclitaxel
Placebo
carboplatin paclitaxel/
nab-paclitaxel
Atezolizumab
carboplatin
nab-paclitaxel
Carboplatin nab-paclitaxel
ORR44.7%35.4%73.0%75.0%50.0%62.6%38.4%49.7%41.0%
mDoR//8.2 m8.6 m4.2 m8.8 m4.9 m7.3 m5.2 m
mPFS5.5 m4.9 m7.6 m7.6 m5.5 m6.4 m4.8 m6.3 m5.6 m
PFS HR0.540.520.48/0.560.71
mOSNANANANANA15.9 m11.3 m14.2 m13.5 m
OS HR0.57/0.640.88
Abbreviations: LUSC, lung squamous cell carcinoma; ORR, objective response rate; mDoR, median duration of response; mPFS, median progression-free survival; HR, hazard ratio; mOS, median overall survival; NA, not available.
Table 4. Summary of PD-L1 immunohistochemistry assays in non-small cell lung cancer (NSCLC).
Table 4. Summary of PD-L1 immunohistochemistry assays in non-small cell lung cancer (NSCLC).
Antibody Clone22C328-8SP142SP263
Species of originMouseRabbitRabbitRabbit
InstrumentDako
Autostainer Link48
Dako
Autostainer Link48
Ventana
Benchmark ULTRA
Ventana
Benchmark ULTRA
Detection systemEnVision FlexEnVision FlexOptiview and
Amplification
Optiview
Targeted cellTCTCTC/ICIC
PD-L1 cutoffTC ≥ 1%TC ≥ 1%TC ≥ 50%/IC ≥ 10%TC ≥ 1%
IndicationPembrolizumab
CDx
Nivolumab
complementary diagnostics
Atezolizumab
CDx
Pembrolizumab CDx, Nivolumab
complementary diagnostics
Abbreviations: PD-L1, programmed death-ligand 1; NSCLC, non-small cell lung cancer; TC, tumor cell; IC, immune cell; CDx, companion diagnostics.
Table 5. Overview of published studies assessing tumor mutational burden (TMB) in LUSC.
Table 5. Overview of published studies assessing tumor mutational burden (TMB) in LUSC.
Cancer TypeNSCLC
(n = 36)
Stage IV or Recurrent NSCLC (n = 312)Stage IV or Recurrent LUSC (n = 100)
Clinical trial/drugAnti-PD-1/PD-L1 monotherapy, anti-CTLA-4/anti-PD-1/PD-L1 combinationsCheckMate 026/nivolumab or platinum-based chemotherapyCheckMate227/nivolumab plus ipilimumab, nivolumab plus chemotherapy, or chemotherapy
Definition of TMBThe number of somatic mutations by NGSThe total number of somatic missense mutations present in a baseline tumor sampleNA
Detection method of TMBFoundationOne assayWhole-exome sequencingFoundationOne CDx assay
Cut-off value of TMB≥20 mutations/Mb≥243 mutations≥10 mutations/Mb
Type of benefitRR, PFS, OSPFS, no benefit of OSORR, PFS
Abbreviations: TMB, tumor mutational burden; LUSC, lung squamous cell carcinoma; NSCLC, non-small cell lung cancer; PD-1, programmed death-1; PD-L1, programmed death ligand 1; CTLA-4, cytotoxic T lymphocyte antigen 4; NGS, next-generation sequencing; NA, not available; RR, response rate; PFS, progression-free survival; OS, overall survival; ORR, objective response rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yuan, H.; Liu, J.; Zhang, J. The Current Landscape of Immune Checkpoint Blockade in Metastatic Lung Squamous Cell Carcinoma. Molecules 2021, 26, 1392. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26051392

AMA Style

Yuan H, Liu J, Zhang J. The Current Landscape of Immune Checkpoint Blockade in Metastatic Lung Squamous Cell Carcinoma. Molecules. 2021; 26(5):1392. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26051392

Chicago/Turabian Style

Yuan, Hong, Jing Liu, and Jun Zhang. 2021. "The Current Landscape of Immune Checkpoint Blockade in Metastatic Lung Squamous Cell Carcinoma" Molecules 26, no. 5: 1392. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26051392

Article Metrics

Back to TopTop