We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Immunotherapy-based combination strategies for treatment of EGFR-TKI-resistant non-small-cell lung cancer

    Yan Chen

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    ,
    Zijun Chen

    Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    ,
    Rui Chen

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    ,
    Cheng Fang

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    ,
    Chu Zhang

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    ,
    Mei Ji

    **Author for correspondence:

    E-mail Address: zlkjimei@163.com

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    &
    Xin Yang

    *Author for correspondence:

    E-mail Address: yangxindoctor@163.com

    Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China

    Published Online:https://doi.org/10.2217/fon-2021-0862

    Abstract

    The rapid development of molecular targeted therapy brings hope to patients with advanced non-small-cell lung cancer (NSCLC). However, drug resistance inevitably occurs during treatment with EGFR-tyrosine kinase inhibitors (TKIs). Osimertinib, a third-generation EGFR-TKI, shows a favorable prognosis in T790M-positive NSCLC. Unfortunately, acquired resistance is still a challenge for both patients and clinicians. There is still no consensus on the optimal treatment. PD-1 and its ligand receptor 1 (PD-L1) inhibitors have yielded great progress, especially in patients with no actionable mutations. In this review, the authors take stock of the relationship between EGFR mutations and PD-L1 expression and summarize the important clinical studies on immunotherapy-inhibitor-based treatment in patients with EGFR-TKI-resistant NSCLC.

    Plain language summary

    Lung cancer is one of the most common malignant cancers worldwide. Specific genes are known to drive cancer growth in advanced non-small-cell lung cancer (NSCLC) and targeted therapies against these genes and their proteins have significantly improved survival. However, resistance to these therapies inevitably occurs and there is still no consensus on the best treatment strategy after resistance develops. Several related articles have discussed the relationship between treatment resistance and the production of PD-L1 proteins in cancer cells (which helps them avoid the body's immune system). EGFR-tyrosine kinase inhibitors (TKIs), a type of targeted therapy, were also reported to influence the production of PD-L1. Therefore, the authors hypothesize that immunotherapy (a type of targeted therapy that forces the body's immune system to attack cancers regardless of PD-L1 production) may be a new option for treatment-resistant patients. In this review, the feasibility of EGFR-TKIs in combination with immunotherapy is clarified.

    Lung cancer is one of the most widely studied malignancies worldwide [1] and can be classified as non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC) on the basis of its histopathological character. NSCLC accounts for approximately 80% of all lung cancer cases [2]. Around 75% of patients are found to have invasion and distant metastasis due to the lack of effective early screening and diagnosis and the 5-year survival rate is less than 15% [3]. The management of advanced NSCLC has recently attracted extensive attention and EGFR-tyrosine kinase inhibitors (TKIs) are considered the standard first-line therapy for patients with NSCLC with EGFR mutations [4,5], in light of significantly improved objective response rates (ORR; 67.0 vs 40.8%) and median progression-free survival (PFS; 10.9 months vs 7.4 months) compared with traditional chemotherapy [6]. Unfortunately, the majority of patients will exhibit disease progression within one year of EGFR-TKI treatment [7]. A meta-analysis found that osimertinib noticeably improved PFS, ORR and disease control rate (DCR), especially in T790M-positive NSCLC compared with other EGFR-TKIs or chemotherapy. Despite the dramatic response to osimertinib, most T790M-positive cases develop resistance within 9–14 months [8]. EGFR-TKI resistance remains a crucial unmet need and the optimal strategy is not yet clearly defined after treatment failure of EGFR-TKIs. Nivolumab and pembrolizumab, a PD-1 antibody, have been authorized for advanced NSCLC for their superior efficacy and tolerable toxicity over traditional chemotherapy by the US FDA [9–11]. However, only about 20% of patients with NSCLC respond to PD-1/PD-L1 monotherapy [12]. The expression of PD-L1 on tumors cells can be upregulated by EGFR activation [13]. EGFR-TKIs were also found to have the potential to affect the tumor microenvironment (TME) [14–16], providing a theoretical basis for the application of immune checkpoint inhibitors (ICIs) in NSCLC with poor efficacy of targeted therapy coupled with upregulated PD-L1 expression. Multiple intensive clinical studies are currently underway to improve the clinical benefit for such patients. The current review provides insights into the recent development of a combination strategy based on immunotherapy for EGFR-TKI-resistant NSCLC.

    EGFR mutation & PD-L1 expression

    Researchers have examined the relationship between EGFR mutation status and the expression of PD-L1 with controversial results. Multiple preclinical studies suggest that PD-L1-positivity in patients with EGFR mutant NSCLC is higher than in those with wild-type EGFR mutations and PD-1 antibody prolonged survival in a nude mouse model [17–19]. Conversely, a negative or insignificant correlation was reported in this setting and PD-L1 was highly expressed in EGFR-wild-type NSCLC with a higher sensitivity to immunotherapy [20–23]. A variety of factors, such as immunohistochemistry (IHC) scoring criteria for PD-L1 in different clinical studies, tumor heterogeneity, baseline characteristics of patients as well as tumor stage and previous treatment before detection may explain such differences between experimental studies [24]. Much work remains to identify the heterogeneity between them. Recent studies indicate that cell-intrinsic EGFR signaling through the induction of T-cell apoptosis and promotion of immune escape in EGFR-mutated NSCLC directly or indirectly upregulated the PD-L1 expression in tumor cells, thus enhancing sensitivity to PD-1/PD-L1 blockade [25]. This regulatory mechanism was first found by Akbay and colleagues in 2013 [26] and has recently attracted attention from scholars. Downregulation of PI3K, AKT and mTOR were confirmed to have the potential to reduce the PD-L1 expression in recent research [27,28], which suggests that EGFR mutations may participate in the regulation of PD-L1 expression by activating the PI3K/AKT/mTOR signaling pathway [29]. Furthermore, a study showed that EGF stimulation, exon 19 deletions and L858R mutations also led to the activation of EGFR and induced PD-L1 expression, which may occur through the mechanism of the p-ERK1/2/p-c-Jun pathway [30]. NF-κB, an important downstream pathway of EGFR activation, can be activated by the tumor suppressor gene and cytokines produced by the inflammatory microenvironment. A study found that subunits of NF-κB could bind to the PD-L1 promoter in NSCLC cells and directly regulate PD-L1 transcription in the form of a complex [31]. In addition, the expression of PD-L1 decreased with silencing of the subunits of NF-κB in tumor cell lines with EGFR mutation, proving that this type of regulation requires the involvement of NF-κB [32]. That the IL-6/JAK/STAT3 pathway participated in the activation of the EGFR signaling pathway had been previously revealed [33]. STAT3, the important regulatory molecule in the downstream signaling pathway of EGFR, can bind to the promoter region of PD-L1 and promote its transcription. Further activation of the EGFR pathway can stimulate the secretion of IL-6 and then activate the JAK/STAT3 signaling pathway [34,35], indicating that the EGFR pathway can upregulate PD-L1 expression through the mechanism of the IL-6/JAK/STAT3 pathway. This hypothesis was subsequently verified in the research by Zhang and colleagues [36]. The MEK/ERK signaling pathway in the MAPK pathway is usually activated by abnormal amplification or mutation of the RTK, and the MEK/ERK pathway exerts a regulatory role in PD-L1 expression in EGFR-mutated NSCLC cells [37,38]. Ota et al. further revealed that both the EML4-ALK fusion gene and mutant EGFR upregulated PD-L1 expression by activating the PI3K-AKT and MEK-ERK signaling pathways in NSCLC [37]. Knowledge of the regulatory mechanisms contributes to the development of innovative therapeutic options to overcome the limited clinical response to EGFR-TKIs. However, the specific molecular mechanism has not been firmly established and additional investigation is required. Figure 1 described the possible mechanisms by which EGFR regulates the expression of PD-L1.

    Figure 1. EGFR regulates the expression of PD-L1 and upregulation of PD-L1 leads to EGFR-tyrosine kinase inhibitors resistance.

    TKI: Tyrosine kinase inhibitor.

    Correlation between PD-L1 expression & EGFR-TKIs

    The epidemiological correlation between PD-L1 expression and activation of EGFR in NSCLC has been observed in many studies. However, the relationship between PD-L1 expression and EGFR-TKIs and the potential mechanism of resistance to this treatment requires more research. Immunophenotypes have the potential to influence the effectiveness of targeted therapy. Recent studies have assessed the relationship between PD-L1 expression and sensitivity to EGFR-TKIs and found that high expression of PD-L1 exhibited poor clinical efficacy characterized by shortened ORR (35.7 vs 67.3%, p = 0.002) and PFS (3.8 vs 9.5 months, p < 0.001) respectively, especially for patients with NSCLC with primary resistance [39], To better explain this suboptimal effect, Lin et al. explored the impact of TME on EGFR-TKIs and found that patients who completed EGFR-TKI therapy exhibited increased infiltration of CD8+ T cells and dendritic cells (DCs) and decreased Foxp3+ Treg cells in the early stage. However, these changes disappeared at the late stage of treatment, and myeloid-derived suppressor cells (MDSCs) continued to increase throughout treatment [40–42]. Notably, Isomoto et al. retrospectively analyzed the TME of 138 patients with EGFR-mutated NSCLC with EGFR-TKI resistance by non-T790M and found lower tumor mutational burden (TMB) FOXP3+ Tregs and higher PD-L1 expression level as well as CD8+ tumor-infiltrating lymphocytes (TILs) densities, which generally confer a promising TME for ICI treatment [16]. Gainor et al. also observed a 20% increase in PD-L1 expression in NSCLC samples with sensitized EGFR mutation [43]. A similar result was found in erlotinib-resistant individuals; the expression of PD-L1 was consistently at a high level [44]. Inomata et al. found that the proportion of PD-L1 strong positive in T790M-negative patients was greater than in those with positive-T790M (p = 0.0565) and those who developed acquired resistance mediated by non-T790M mechanism also showed a superior PFS during subsequent immunotherapy than those with positive-T790M (16.5 vs 8.6 months, p = 0.001) [45]. A subsequent retrospective study also reported a better PFS in patients with a high expression of PD-L1 after treatment with EGFR-TKIs (7.1 vs.1.7 months, p = 0.0033) [46]. Such dynamic changes associated with EGFR-TKI agents indicate that a subset of patients with high-PD-L1 NSCLC who failed treatment with EGFR-TKIs might be more suitable for immunotherapy. Notably, a higher positive rate (TPS: ≥1%) of PD-L1 in a subset of patients with NSCLC harboring EGFR activation was also demonstrated to be associated with primary resistance to EGFR-TKIs when compared with those with lower PD-L1 expression (45.5 vs 12.3%, p < 0.001) by the mechanism upregulating the expression of YAP1, the transforming growth factor/Drosophila mothers against decapentaplegic (TGF-Smad) signaling pathway and continuous activation of the extracellular regulated ERK pathway by the PD-L1/BAG-1 axis [13,40,47–49]. EGFR-TKIs were also found to downregulate the PD-L1 expression of tumor cells through the IL-6/JAK/STAT3 pathway and Hippo/YAP signaling pathway [36,50]. Such studies suggest that in addition to targeting the EGFR signaling pathway to attenuate tumor proliferation and improve survival, downregulation of PD-L1 expression and consequent stimulation of anti-tumor immune effects also have the potential to overcome EGFR-TKI resistance in NSCLC with strong PD-L1 expression, especially for those negative for T790M. Knowledge of identified primary or acquired resistance mechanisms continues to accumulate, motivating the development of diverse new therapies to overcome EGFR-TKI resistance.

    Immunotherapy monotherapy

    Research on the curative efficacy of immunotherapy in EGFR-TKI-resistant NSCLC has yielded controversial results and the effect of ICIs in patients who failed EGFR-TKIs has not been clarified. Regardless of the dramatic success of immunotherapies in some patients with lung adenocarcinoma, patients with EGFR-mutated NSCLC continue to be excluded from some critical phase III randomized controlled trials (RCTs) of first-line immunotherapy due to suboptimal response [51,52]. The activity of immunotherapy in patients with PD-L1-positive EGFR-mutant NSCLC has been described in previous clinical trials. The results from the subgroup of the Checkmate057 trial, which analyzed 82 patients with EGFR-mutated NSCLC who completed nivolumab after the failure of EGFR-TKIs, indicated no dramatic response to nivolumab compared with docetaxel (HR: 1.18, 95% CI: 0.69–2.00) in terms of PFS and overall survival (OS) benefit [53]. The KEYNOTE010 trial aimed to evaluate second-line treatment with pembrolizumab and docetaxel for PD-L1 positive NSCLC (TPS ≥1%). In a subgroup of 86 advanced patients previously treated with EGFR-TKIs, no significant OS benefit was demonstrated compared with the docetaxel (HR: 0.88, 95% CI: 0.45–1.70) [54]. Similarly, the latest results of the WJOG8515L trial presented at the American Society of Clinical Oncology (ASCO) conference in 2021 indicated that the therapeutic effect of nivolumab was much inferior to chemotherapy (ORR: 9.6 vs 36%; DCR: 38.4 vs 76%; median PFS [mPFS]: 1.7 months vs 5.6 months) regardless of PD-L1 expression [55]. Consistently with previous results from the recent POPLAR and OAK trials, patients with EGFR-mutated NSCLC with previous EGFR-TKI treatment derived no OS benefit from PD-1 inhibitors. It is crucial to underly the mechanisms of this unfavorable effectiveness with immunotherapy in the EGFR-mutated subset. TME, consisting of many indicators such as PD-L1 expression, TMB and Tregs has been revealed to influence the response of immunotherapy in patients with EGFR-positive NSCLC [40,56]. Preliminary studies showed that EGFR-signaling cancer cells may overexpress immune suppressors, such as TGF-β and IL-10 cytokines, which may directly restrain the capacity of natural killer (NK) cells, further inhibit the recruitment of effector CD8+ T cells and facilitate Treg infiltration, finally leading to the proliferation of tumor cells [57,58]. High levels of PD-L1 expression and TMB are considered promising predictive biomarkers for immunological therapy efficacy based on recent studies [59,60]. EGFR mutations were strongly associated with T-cell depletion and dormancy immunophenotypes (high expression of CD3, low expression of Ki67 and granase B) compared with wild-type EGFR, which may result in impaired TIL-mediated anti-tumor effects [61].

    Evidence also indicates that CD73 is overexpressed in EGFR-mutated NSCLC, which can split ATP into adenosine and act on A2a/A2b receptors to exert an immunosuppressive effect. It can also negatively regulate the function of DCs and NK cells, polarize macrophages to M2 and inhibit the anti-tumor response mediated by T cells, thus enhancing immune escape, metastasis and the proliferation of tumors and activating immunosuppressive effect by activating Treg cells and MDSCs [62,63]. In addition, Gainor et al. found only 11.6% of EGFR-TKI-progressed tumors had concurrent high levels of PD-L1 expression and CD8+ TILs [43]. The little available evidence may, to some extent, explain the discouraging clinical response to ICIs among NSCLC patients with sensitizing EGFR mutations [64,65]. Despite these unfavorable subgroup analyses in RCTs, modest but promising efficacy was initially verified in some preclinical studies. The ATLANTIC study explored the efficacy and safety of durvalumab as third-line treatment in EGFR-positive NSCLC pre-treated with EGFR-TKIs (PD-L1 expression ≥25%), and encouraging clinical data were observed in terms of the ORR (12.2%, 95% CI: 5.7–21.8) [66]. Jia et al. investigated dynamic TME responding to EGFR-TKIs using a murine model and the results showed that tumor cells with the EGFR mutation responded to EGFR-TKIs as early as the first day, accompanied by significant tumor shrinkage. However, no significant change in the anti-tumor effect was observed compared with the untreated group after long-term EGFR-TKI infiltration of CD8+ T cells and macrophages were significantly lower in EGFR-TKI-resistant cells [25].

    EGFR-mutated NSCLC represents a heterogeneous subgroup of patients. Recent studies have found that different subtypes of EGFR mutations derive a differential clinical advantage from ICIs [67]. Patients with EGFR L858R mutation showed a high TMB and significantly improved ORR compared with those with EGFR exon 19 deletion [68]. Insertions in exon 20 (Ex20ins) of EGFR were also related to greater benefit with comparable PFS and ORR to immunotherapy compared with those with common EGFR and HER2 mutations (PFS: 4.0 vs 1.9 vs 1.9 months; ORR: 19 vs 0 vs 9%; p < 0.05), which may be partially explained by relatively higher PD-L1 expression in individuals with Ex20ins mutation [69]. EGFR mutated individuals usually establish an immunosuppressive noninflammatory TME leading to resistance to immunotherapy. Nevertheless, patients with EGFR mutations are not forbidden from immunotherapy; highly selected patients with high expression of immunological biomarkers (PD-L1 and TMB) are more likely to obtain benefits from immunotherapy. The discouraging preliminary clinical results emerging from early-phase trials warrant additional long-term follow-up and further investigation requires carefully estimating the effects and safety profile of ICIs in EGFR-TKI-resistant NSCLC, including specific duration of treatment (DOR), and subtype of EGFR mutations. In the near future, outcomes from numerous ongoing clinical trials evaluating immunotherapy for EGFR-TKI-resistant populations will be reported.

    EGFR-TKI–immunotherapy combination treatment

    Targeted therapies may reduce tumor-mediated immunosuppression by eliminating the production of tumorigenic inflammation and suppressing immunosuppressive cell types indicating a synergistic effect with immunotherapies that enhances T-cell immune response [70,71]. Preclinical studies also found that EGFR-TKIs were indirectly capable of inhibiting the function of Treg cells through the EGFR/GSK-3/Foxp3 axis and strengthening the anti-tumor effect by inducing tumor-specific T cells to release inflammatory cytokines such as IFN-γ, thereby significantly increasing peripheral NK cells and IFN-γ, and the level of IL-6 decreased after EGFR-TKIs [30,72]. Short-term exposure to low-dose EGFR-TKIs (e.g., erlotinib) can make tumor cells tend toward an epithelial phenotype and enhance the recognition and lysis of tumors modulated by antigen-specific T cells and NK cells [73]. Recent studies also showed that activation of the EGFR pathway may inhibit immune responses in murine melanoma models by activating Treg cells or decreasing T-cell chemotactic levels [40], while gefitinib is capable of reversing the immunosuppression effect by upregulating the major receptors (NKG2D) expression and enhancing immune recognition of NK cells, especially for those resistant to common mutations (EGFR L858R + T790M) [74]. The indirect and remarkable enhancement of anti-tumor immunity after EGFR-TKIs reverses immunosuppression in the TME and makes it possible for combinations of targeted therapy and immunotherapies. Immunotherapy combined with EGFR-TKIs is under scrutiny in current clinical studies, and the effectiveness and feasibility of this exploratory regime remain to be confirmed. The subgroup of KEYNOTE-012 analysis indicated that the combination of pembrolizumab and erlotinib provided better clinical benefits for EGFR-TKI-naive NSCLC individuals regarding moderate ORR (41.7 vs 14.3%) and prolonged mPFS (19.5 months vs 1.4 months) without dose-limiting toxicity or grade 5 adverse events (AEs) compared with those treated with pembrolizumab and gefitinib [75]. However, the efficacy of the combination therapy requires clarification with further phase II clinical trial data due to lack of an adequate sample population.

    Recently, the simultaneous introduction of various strategy profiles into standard practice has led to clinical challenges regarding optimal treatment sequencing and unintended serious toxicities. The CAURAL trial compared osimertinib with durvalumab in EGFR-TKI pretreated participants with T790M positive mutation. The results showed no historically improved ORR (64 vs 80%), DOR (17.5 vs 21.4 months) or DCR (93 vs 100%) in the combined treatment group compared with those treated with osimertinib monotherapy and the trial was terminated early due to the high incidence of interstitial lung disease (38%) [76]. Several concerns have been raised regarding the potential high incidence of AEs associated with immunotherapy combined with EGFR-TKIs in patients who progressed on EGFR-TKIs. A meta-analysis of PD-1/PD-L1 blockade in combination with EGFR-TKIs showed an ORR of 55% (95% CI: 28–83%), the incidence of grade 3–5 drug-related AEs (DRAEs) was 41% (95% CI: 20–62%), of which the most common incidence was pneumonia at 3% (95% CI: 1–7%), and drug-related mortality was 1% (95% CI: 3–6%) [77]. Similar safety profiles were also reported in 13 patients with EGFR-TKI-naive NSCLC with positive EGFR included in the CheckMate370 (NCT02574078) phase II/III RCT of nivolumab and crizotinib, showing that only five cases (38%) had achieved partial response (PR), and the strategy was ultimately discontinued due to severe hepatotoxicity (38%) [78]. Consistently with previous reports, nivolumab in combination with erlotinib was associated with 19% grade 3/4 toxicity [79] and the incidence of grade 3/4 elevation of liver enzymes in combination with duvalizumab was high (40–70%). Moreover, 39% of the patients who received atezolizumab and erlotinib experienced grade 3/4 immune-related AEs (irAEs) [71]. Despite a promising ORR (70%) for EGFR-TKI-naive patients from the combination of durvalumab and osimertinib in the TATTON trial, more attention should be paid to the high incidence of grade 3/4 irAEs (34% of the enrolled patients developed interstitial pneumonia) [80]. Results from the current study showed that 15% (6/41) of all patients receiving sequential osimertinib with PD-1/PD-L1 inhibitors developed severe irAEs. In contrast, no severe irAEs were observed in those who received osimertinib sequentially with PD-1/PD-L1 inhibitors (0/29) or PD-1/PD-L1 inhibitors sequentially with other EGFR-TKIs (afatinib or erlotinib, 0/27) [81]. The results highlight the urgent need for careful consideration in the selection of initial treatment for patients with NSCLC, particularly for osimertinib in patients who have recently been treated with PD-1/PD-L1 inhibitors. Most studies suggest that the joint application of PD-1/PD-L1 inhibitors and EGFR-TKIs can improve clinical ORR and prolong PFS. However, there were also many serious AEs, especially interstitial pneumonia. Given the challenge of the potential toxicity of combination therapy, which resulted in the early closure of some studies (and the results from these trials are preliminary and unfavorable), the optimal sequence, time interval and dosage of administration remain unclear. At present, multiple phase I clinical trials are intensively investigating the efficacy and safety of this combined therapy, including NCT01454102, NCT02039674 and NCT02013219, which are underway to estimate the combination of EGFR-TKIs and nivolumab, pembrolizumab and atezolizumab, respectively. Table 1 shows relevant clinical trials that assess the combination of anti-PD-1/PD-L1 therapy and EGFR-TKIs in patients with NSCLC resistant to EGFR-TKIs.

    Table 1. Ongoing studies of PD-1/PD-L1 inhibitors plus EGFR-tyrosine kinase inhibitors in EGFR-tyrosine kinase inhibitor-resistant non-small-cell lung cancer.
    Trial (NCT identifier)EGFR-TKIImmune checkpoint inhibitorPatientsnPhaseStatus
    NCT02364609 (2015)AfatinibPembrolizumabNSCLC resistant to erlotinib11ICompleted
    NCT02040064 (2014)GefitinibTremelimumabEGFR-mutant NSCLC27ICompleted
    NCT02630186 (2015)RociletinibMPDL3280A (anti-PD-L1 antibody)Progressed after prior first- or second-generation EGFR-TKIs3IITerminated
    CAURAL/NCT02454933 (2015)OsimertinibDurvalumabNSCLC with progression after EGFR-TKIs, T790M positive344IActive, not recruiting
    TATTON/NCT02143466 (2014)OsimertinibDurvalumab or durvalumab + tremelimumabNSCLC with progression after EGFR-TKIs, T790M positive344IActive, not recruiting
    NCT01454102 (Checkmate 012) (2011)ErlotinibNivolumabAdvanced NSCLC with EGFR mutation with EGFR-TKI resistance545IActive, not recruiting
    NCT02039674 (2014)Erlotinib or gefitinibPembrolizumabEGFR-mutant NSCLC progressed following EGFR TKI therapy267I/IICompleted
    GEFTREM/NCT02040063 (2014)GefitinibTremelimumabTKI-resistant EGFR-mutated NSCLC27ICompleted
    NCT01998126 (2013)ErlotinibNivolumab/ipilimumabStages II-4 TKI-naive or TKI-treated EGFR-mutated NSCLC14ICompleted

    TKI: Tyrosine kinase inhibitor; MPDL3280A: Atezolizumab; NSCLC: Non-small-cell lung cancer.

    All data come from ClinicalTrials.gov, accessed November 2021.

    Chemotherapy-immunotherapy combination treatment

    The standard first-line treatment of pembrolizumab in combination with platinum-based chemotherapy is currently approved for patients with nonsquamous cell carcinoma without EGFR/AKL mutations by the US FDA [82]. Numerous studies have suggested that patients with NSCLC with actionable mutations have a poor response to ICIs [43,83], and immunotherapy in combination with chemotherapy is more applicable to NSCLC patients with PD-L1 expression ≥50% [84–86]. Nevertheless, research on cytotoxic chemotherapeutic agents that may affect the immune microenvironment in terms of PD-L1 expression, TMB and activity of anti-tumor immunity has been elucidated [87]. Studies have shown that chemotherapeutic agents, such as gemcitabine, paclitaxel and carboplatin, can induce the upregulation of PD-L1 by activating the NF-κB signaling pathway [88]. In addition, the immunomodulatory property of chemotherapy can expose or release neoantigen molecules in tumor cells and transform them into immunogenic cells [89]. This process eventually results in a series of immune reactions, such as the activating of DCs through toll-like receptors (TLRs), the enhancement of effector T cells, reduction of Treg and MDSCs, activation of mature DC cells and recruitment of related antigen-presenting cells to kill tumors and facilitate tumor immune response [90–92]. Remarkably, chemotherapy contributes to significant activation of anti-tumor immunity and an ICI-based strategy may extend this synergistic effect [93]. A pooled analysis of three RCTs (KEYNOTE-021, KEYNOTE-189 and KEYNOTE-407) showed that combined pembrolizumab and standard-care chemotherapy considerably improved OS (HR: 0.63, 95% CI: 0.50–0.79) and PFS (HR: 0.68; 95% CI: 0.56–0.83) over chemotherapy alone with manageable AEs [94]. Additionally, exposure to single-agent chemotherapy after treatment with EGFR-TKIs yielded improved response rates compared with pre-immunotherapy in patients with advanced NSCLC [95]. It is thus hypothesized that chemotherapy in tandem with immunotherapy may improve the anti-tumor response. The preliminary results of a single-arm, multicenter, phase II study of toripalimab and carboplatin/pemetrexed in EGFR-positive patients who failed prior EGFR-TKIs were presented at the CSCO conference in 2019. A total of 40 eligible patients were treated with toripalimab (a PD-1 inhibitor) and pemetrexed/carboplatin for four or six cycles (every 3 weeks), followed by a maintenance combination regimen until disease progression. The encouraging results showed that 20 patients achieved PR (50%), 15 patients achieved stable disease (SD; 37.5%), the ORR was 50%, DCR reached 87.5%, median PFS was 7.0 months and the mPFS of PD-L1 positive patients reached 8.2 months. The results indicated that the combination regimen was superior to chemotherapy alone both in PFS and ORR in the PD-L1 positive subset [96]. Subsequently, the clinical data from the EGFR-positive (n = 124) subgroup of the IMPOWER150 trial showed that atezolizumab plus bevacizumab plus carboplatin plus chemotherapy (ABCP) had historically increased median OS compared with bevacizumab plus carboplatin plus paclitaxel (BCP; 29.4 months vs 18.1 months, HR: 0.6, 95% CI: 0.31–1.14), and the group with baseline liver metastasis also achieved parallel results [97]. Successful results from research to date support the potential of the ABCP regimen as an innovative therapeutic option for post-line treatment after failure on EGFR-TKIs.

    The application of consolidation therapy with durvalumab after concurrent chemoradiotherapy (CRT) in unresectable stage III NSCLC remains controversial. A retrospective analysis explored the PFS with durvalumab in this setting and no improvements were observed compared with those who completed CRT alone. Patients experienced a high frequency of severe irAEs, numerically similar to the subset of analysis from the PACIFIC trial [98]. To extend survival benefit and attenuate the occurrence of resistance to targeted therapy, more and more clinical trials are in the process of investigating the feasibility of chemotherapy and immunotherapy combination treatment [99]. The ongoing clinical trials KEYNOTE-789 (NCT03515837), CheckMate722 (NCT02864251) and Orient-31 (NCT03802240) will explore the response to ICIs plus traditional chemotherapy after progression on EGFR-TKIs. Table 2 summarizes relevant clinical trials that evaluate the combination of ICIs and chemotherapy in patients with NSCLC after EGFR-TKI treatment failure.

    Table 2. Ongoing studies of PD-1/PD-L1 inhibitors plus chemotherapy in EGFR-tyrosine kinase inhibitor-resistant non-small-cell lung cancer.
    Trial (NCT identifier)Immune checkpoint inhibitorsChemotherapyPatientsnPhaseStatus
    NCT04322890 (2020)PD-1 antibodyChemotherapyTKI-resistant EGFR-mutated NSCLC100NARecruiting
    NCT04405674 (2020)Tislelizumab (anti-PD-1 antibody)Carboplatin/pemetrexedTKI-resistant EGFR-mutated NSCLC66IIRecruiting
    NCT03515837 (KEYNOTE-789) (2018)PembrolizumabPemetrexed + platinumTKI-resistant EGFR-mutated NSCLC492IIIActive, not recruiting
    NCT03513666 (JS001) (2018)ToripalimabPemetrexed + carboplatinEGFR mutation-positive and T790M (-) after progression on EGFR-TKIs40IIActive, not recruiting
    NCT04147351 (2019)Atezolizumab+ bevacizumabCarboplatin or cisplatin + pemetrexedEGFR-mutant metastatic NNSCLC, failed EGFR-TKIs22IIRecruiting
    NCT03802240 (ORIENT-31) (2019)SintilimabPemetrexed + cisplatinAdvanced or metastatic NSCLC, failed EGFR-TKIs600IIIRecruiting
    WJOG8515L (2017)NivolumabCarboplatin + pemetrexedEGFR mutation-positive NSCLC with resistance to EGFR-TKIs due to mechanisms other than T790MNAIIOngoing
    NCT02864251 (Checkmate722) (2016)Nivolumab or ipilimumabChemotherapyNSCLC with EGFR mutation, failed first- or second-generation EGFR-TKIs365NAActive, not recruiting
    ILLUMINATE (NCT03994393) (2019)Durvalumab +tremelimumabPlatinum + pemetrexedEGFR mutation-positive NSCLC progressed following EGFR-TKIs100IIRecruiting
    JS001/NCT03513666 (2018)ToripalimabCarboplatin + pemetrexedEGFR mutation-positive and T790M negative NSCLC after progression on EGFR-TKIs40IIActive, not recruiting
    NCT04971187 (2021)Bintrafusp Alfa (anti-PD-1 antibody)Platinum-based chemotherapyEGFR-TKI-resistant NSCLC with EGFR mutation40IIRecruiting
    NCT04405674 (2020)Tislelizumabs (anti-PD-1 antibody)Carboplatin + nab-paclitaxel or pemetrexedEGFR-TKI-resistant NSCLC with EGFR mutation66IIRecruiting
    NCT04970043 (2021)CamrelizumabPemetrexed + carboplatinEGFR-TKI-resistant NSCLC with EGFR mutation58IINot yet recruiting
    PIKACHU/ NCT04322890 (2020)PD-1 antibodyChemotherapyTKI failure, EGFR/ALK/ROS1 mutation-positive advanced NSCLC100Recruiting

    TKI: Tyrosine kinase inhibitors; NA: Not available; NSCLC: Non-small-cell lung cancer.

    All data come from ClinicalTrials.gov, accessed November 2021.

    Antiangiogenic-immunotherapy combination treatment

    VEGF, a highly specific provascular endothelial cell growth factor, promotes the movement, proliferation and division of vascular endothelial cells [100], increases vascular permeability and has proangiogenic activity [101]. It can inhibit the maturation of DCs and the response of effector T cells and increases the infiltration of MDSCs, Tregs and monocytes/macrophages, contributing to the infiltration of various immunosuppressive cells into tumor tissues and exerting an immunosuppressive role, which may be stimulated by EGFR/ALK signaling [102]. The relationship between VEGF and its receptors and the immune system is complex. Antiangiogenic drugs can activate the immune system and immunotherapy can also combat angiogenesis [103–105], providing a novel and powerful rationale for the combination of anti-VEGF and ICI therapies. Previous in vitro experiments revealed that a combination of low-dose apatinib and PD-L1 blockade showed an obvious anti-tumor effect in a syngeneic lung cancer mouse model [106]. Wu et al. [107] also showed that anti-PD-1 in combination with endostar (antivascular drug) can remarkably attenuate tumor growth by inhibiting the accumulation of MDSCs in a mice model. Interestingly, recent evidence suggested that the combined strategy of PD-1-based immunotherapy and antiangiogenic drugs (e.g., endostar) synergistically stimulate the immune response of tumor cells and dramatically enhance the anti-tumor effect by suppressing the common PI3KAKT/mTOR pathway in an LLC mouse model [107]. These preclinical studies corroborated the complementary therapeutic effects of this combination. Additionally, the synergistic benefit of the combination of antiangiogenic therapy with ICIs has been reported in an observational study [108]. As early as the ASCO conference, 2019, results on the combination of camrelizumab and apatinib have been announced with a 30.8% ORR and 5.9 months median PFS [109]. A recent study of 69 patients with NSCLC receiving anti-PD-L1 agents (nivolumab or pembrolizumab) in combination with antiangiogenic therapy are presented and the short-term outcomes include a moderate ORR of 31.9% (95% CI: 20.6–43.2%), and a median PFS of 8.37 months (95% CI: 1.0–9.5) [110]. Albina et al. [111] reported that the PFS of a female patient resistant to erlotinib and osimertinib was significantly improved after combination therapy consisting of carboplatin, paclitaxel, atezolizumab and bevacizumab. However, it is unclear whether a combination of anti-PD-1 antibody and antiangiogenic therapy can bring more survival benefits for the EGFR-TKI-resistant NSCLC population. A subsequent clinical case reported a stage IA lung adenocarcinoma patient with exon 19 deletion mutations after first-line chemotherapy failure who then received gefitinib treatment. After the failure of gefitinib, the patient received immunotherapy in combination with antiangiogenic agents (sintilimab + bevacizumab). Radiological imaging subsequently demonstrated a reduced adrenal metastatic lesion 1 month later, and the PFS was 6 months [112]. The available evidence suggests that a combination of antiangiogenic therapy and immunotherapy displays promising efficacy in EGFR-TKI-resistant NSCLC individuals, particularly in those with high VEGF levels, and more clinical trials are required to confirm these synergistic effects. Table 3 shows relevant clinical trials assessing combined treatment with ICIs and antiangiogenic agents in patients with NSCLC.

    Table 3. Ongoing studies of PD-1/PD-L1 inhibitors plus antiangiogenic treatment in EGFR-tyrosine kinase inhibitor-resistant non-small-cell lung cancer.
    Trial (NCT identifier)Immune checkpoint inhibitorsAntiangiogenicPatientsnPhaseStatus
    NCT03765775 (2018)SintilimabAnlotinibNSCLC with first-generation EGFR-TKI drug resistance, T790M-negative20IIRecruiting
    NCT04116918 (2019)JS001AnlotinibEGFR-TKI-resistant and T790M (-) NSCLC100NARecruiting
    NCT03802240 (2019)SintilimabIBI305EGFR-mutant advanced or metastatic NSCLC with EGFR-TKI resistance600IIIRecruiting
    NCT04120454 (2019)PembrolizumabRamucirumabEGFR mutant recurrent or metastatic NSCLC34IINot yet recruiting
    NCT04245085 (2020)AtezolizumabBevacizumab + chemotherapyEGFR-mutant NSCLC with acquired resistance95IIRecruiting
    NCT03083041 (2017)SHR-1210 (anti-PD-1 antibody)ApatinibEGFR-mutant advanced NSCLC with EGFR-TKI resistance118IIRecruiting
    NCT04316351 (2020)ToripalimabAnlotinib + pemetrexedT790M positive NSCLC (IIIB/ IV) after osimertinib resistance60IIRecruiting
    NCT05078931 (2021)PembrolizumazLenvatinib (anti-VEGF)PD-L1 positive TKI-resistant NSCLC35IINot yet recruiting
    NCT04517526 (2020)Platinum-based chemotherapy + durvalumabBevacizumabRefractory EGFR-mutated stage IV NSCLC60IINot yet recruiting
    NCT04790409 (2021)SintilimabAnlotinibNSCLC with platinum-containing systemic chemotherapy and/or TKI failure30IIRecruiting
    NCT04147351 (2019)Platinum and pemetrexed + atezolizumabBevacizumabEGFR-mutated NSCLC after failure of EGFR-TKIs22IIRecruiting

    TKIs: Tyrosine kinase inhibitors; IBI305: Bevacizumab; JS001: Toripalimab; NA: Not available; NSCLC: Non-small-cell lung cancer; SHR-1210: Camrelizumab.

    All data come from ClinicalTrials.gov, accessed November 2021.

    Combination CTLA-4 inhibitor-immunotherapy treatment

    PD-1, an immune suppressor transmembrane protein, is found to be not only widely expressed in activated T-lymphocytes, B-lymphocytes, NK cells, monocytes and other immune cells, but also in some tumor cell lines or tumor cell surfaces [113]. It can also exert an important role in promoting tumor immune escape and tumor cell growth by combining with its ligand, PD-L1 [114]. Similar to PD-1, CTLA-4 is another coinhibitory molecule of the B7 family that can relieve the immunosuppressive effect of Treg cells in the TME by blocking the binding of CTLA-4 and B7 molecules on Treg cells, and strengthen the activity of effector T cells [115]. In contrast to the fact that CTLA-4 principally restricts T-cell response in the early immune response in lymphoid tissues, PD-1 mainly inhibits T-cell response in late peripheral tissues, which can contribute effector T cells to long-lasting anti-tumor activity by blocking the binding of PD-L1 of tumor cells to B7-1 on the surface of T cells [116,117]. The response to a single-agent with CTLA-4 inhibitors or PD-1/PD-L1 blockade remains unsatisfactory compared with the combined regime [118]. The joint blocking of PD-1 and CTLA-4 increased tumor infiltration by CD8+ T cells and CD4+ T cells, as well as the ratio of CD8+ and Treg cells, which has been recognized as a complementary mechanism of ICIs [119]. Therefore, it is hypothesized that the combined immunotherapies of CTLA-4 inhibitors and anti-PD1/PD-L1 agents may generate a coadjutant anti-tumor effect compared with single-agent treatment through affecting the complementary pathways in the cancer-immunity cycle [120,121].

    The combination of nivolumab and ipilimumab was approved as applicable for advanced unresectable melanoma [122]. Subsequently, the synergistic effect was verified in a subset population with positive EGFR mutation in the CheckMate-012 trial, and the ORR of such dual immunotherapy with nivolumab and ipilimumab reached 50% [123]. Moreover, the activity of nivolumab plus low-dose ipilimumab as first-line standard care for 288 untreated patients with advanced NSCLC was evaluated in the CheckMate 568 trial [124]. Effective outcomes emerging from the study includ greater ORR in those with PD-L1 expression ≥1% than lower PD-L1 expression (41 vs 15%) [124] and superior PFS in patients with higher TMB than those with low TMB (7.1 vs 2.6 months). In this study, only 29% of patients exhibited grade 3/4 irAEs [124]. Furthermore, the KEYNOTE-021 trial evaluated the combination of pembrolizumab (2 mg/kg) and ipilimumab (1 mg/kg) in previously treated advanced NSCLC with a considerable ORR (30%), although there was no statistical significance (p = 0.0858) [125]. The combined agents of durvalumab (1 mg/kg) and tremelimumab (1 mg/kg), to a great extent, improved OS in an Asian advanced NSCLC population compared with chemotherapy (17.7 vs 10.6 months, p < 0.05), and clinical benefits have been revealed in the post hoc analysis of the MYSTIC trial (multicenter, open-label, phase III) [126]. The aforementioned critical clinical model of combined CTLA-4 and PD-1/PD-L1 inhibitors enhanced the anti-tumor activity of EGFR-TKI-resistant NSCLC, especially for those with higher TMB and PD-L1expression. Although, to date, there have been limited trials on anti-PD-1 and CTLA-4 immunotherapy in EGFR-TKI-resistant NSCLC, which may be attributed to previous cases in which immunotherapy may have a limited effect against NSCLC with EGFR mutations. Therefore, future studies should verify the safety and efficacy of dual immunosuppressive agents in NSCLC with EGFR-TKI resistance. Ongoing phase III trials are evaluating nivolumab plus ipilimumab and durvalumab plus tremelimumab in patients with previously untreated NSCLC [127]. Table 4 summarizes clinical trials that evaluate the combined regime of PD-1/PD-L1 and CTLA-4 inhibitors in patients with advanced NSCLC.

    Table 4. Ongoing studies of PD-1/PD-L1 inhibitors plus CTLA-4 inhibitors in advanced non-small-cell lung cancer.
    Trial (NCT identifier)TreatmentPatientsnPhaseStatus
    NCT03001882 (2016)Nivolumab + ipilimumabTreatment-naive stage IV or recurrent NSCLC232IIActive, not recruiting
    NCT03469960 (2018)Nivolumab + iIpilimumabPD-L1-positive stage IV NSCLC1360IIIRecruiting
    NCT01454102 (2011)Nivolumab + ipilimumabStage IIIB/IV NSCLC472ICompleted
    NCT03515629 (2018)
    NCT03580694 (2018)
    REGN4659 (anti-CTLA-4 monoclonal antibody) + cemiplimabAdvanced or metastatic NSCLC17IITerminated
    NCT03515629 (2018)REGN2810 (anti-PD-1 antibody) + ipilimumabAdvanced or metastatic NSCLC with tumors expressing PD-L1 ≥50%5IIICompleted
    NCT03285321 (2017)Nivolumab + ipilimumabUnresectable stage IIIA/IIIB NSCLC108IIRecruiting
    NCT03262779 (2017)Nivolumab + ipilimumabAdvanced NSCLC resistant to anti-PD-1-axis therapy20IIActive, not recruiting
    NCT03527251 (2018)Ipilimumab + SHR-1210 (anti-PD-1 antibody)Recurrent or metastatic NSCLC10IUnknown
    NCT02869789 (2016)Nivolumab + ipilimumabNewly diagnosed stage IV NSCLC1036IVActive, not recruiting
    NCT03302234 (2017)Pembrolizumab + ipilimumabStage IV, metastatic NSCLC (PD-L1 positive, TPS ≥50%)568IIIActive, not recruiting

    NSCLC: Non-small-cell lung cancer; SHR-1210: Camrelizumab; TPS: Tumor Proportion Score.

    All data come from ClinicalTrials.gov, accessed November 2021.

    Conclusion

    Collectively, this review pointed out that an immunotherapy-based combination regimen might become another selective therapy after the failure of targeted drug therapy by analyzing the close relationship between EGFR mutation and PD-L1 expression status, as well as the dynamic changes of TME of before and after treatment of EGFR-TKIs. At present, drug resistance of EGFR-TKIs is being continuously studied in depth. Meanwhile, it is particularly necessary to explore the resistance mechanism to targeted therapy, additional potential targets, and the development of effective biomarkers to screen the advantaged populations receiving immunotherapy for the realization of precision therapy in the future. Therefore, more research results are required to provide the best-individualized treatment for EGFR-TKI-resistant patients.

    Future perspective

    EGFR-TKIs targeting EGFR signaling pathways have brought favorable survival benefits to the population with EGFR mutation, but most people inevitably develop drug resistance within about one year of treatment. The lack of available treatment options makes the management of such patients challenging. Thus, it is urgent and necessary to investigate innovative strategies. Recently, ICIs have yielded dramatic clinical responses for patients with NSCLC. However, preclinical data showed that EGFR-positive NSCLC patients have a suboptimal response to immunotherapy. However, current clinical experiences of immunotherapy in patients with EGFR-TKI-resistant NSCLC remain absent, and the results of different clinical trials are controversial (Table 5). Given the potential advantages of ICIs in the EGFR-TKI-resistant population, it is vital to understand the correlation between tumor immune-mediated treatment-related biomarkers and tumor drivers in NSCLC. The limited efficacy of immunotherapy in patients with EGFR-positive NSCLC has been described in early-phase trials and different subtypes of EGFR mutations, different administration sequence, time and dosage influence the effect of immunotherapy in this setting, indicating that the strict selection of a high-standard population would be conducive to improving the outcomes of immunotherapy.

    Table 5. Available data on PD-1/PD-L1 inhibitor-based combination therapy in EGFR-tyrosine kinase inhibitor-resistant non-small-cell lung cancer.
    Trial/authorBaselinePatients (n)TreatmentORR (%)Median PFS (months)Median OS (months)PhaseSafetyRef.
    Haratani 2017TKI-pretreated T790M (-)17Nivolumab242.1NARetrospective studyTolerable[15]
    TKI-pretreated EGFR T790M (+)8Nivolumab131.3NA
    ATLANTIC 2018 (NCT02087423)Pretreated EGFR (+)/ALK (+)111Durvalumab12.21.913.32Tolerable[66]
    Keynote-001 2019 (NCT01295827)TKI-pretreated EGFR (+)26Pembrolizumab41.941Tolerable[54]
    Gettinger 2019TKI-pretreated EGFR (+)20Nivolumab + erlotinib155.118.71Tolerable[79]
    GEFTREM (NCT02040064)TKI-pretreated EGFR (+)18Gefitinib + tremelimumab50–80NANA1Tolerable[128]
    TATTON 2020 (NCT02143466)TKI-pretreated EGFR T790M (-)9Osimertinib + durvalumab67NANA1bSevere[80]
    TKI-pretreated EGFR T790M (+)14Osimertinib + durvalumab21NANA
    CAUREL 2018 (NCT02454933)TKI-pretreated EGFR T790M (+)12Osimertinib + durvalumab64NANA3Severe[76]
    TKI-naive EGFR (+)4Osimertinib + durvalumab505.318.6
    IMpower 150 2019Chemotherapy naive EGFR (+)34ABCP7110.2NE3Tolerable[97]
    Chemotherapy naive EGFR (+)45BCP366.921.4
    Zhang 2019TKI-pretreated EGFR T790M (-)40Toripalimab + chemotherapy7 –1Tolerable[96]
    Yang 2019Osimertinib resistance11Nivolumab or pembrolizumab + chemotherapy45.57.47NA1Tolerable[129]
    KEYNOTE-021 cohorts D and H 2018TKI-pretreated EGFR36Pembrolizumab + ipilimumab304.110.92Tolerable[125]

    ABCP: Atezolizumab plus bevacizumab plus carboplatin plus paclitaxel; BCP: Bevacizumab plus carboplatin plus paclitaxel; CT: Chemotherapy; NA: Not available; NE: Not estimable; ORR: Objective response rate; OS: Overall survival; PFS: Progression-free survival rate.

    Although the response to ICIs in EGFR-mutant individuals was worse in those with wild-type EGFR mutation, the high expression of PD-L1 in such patients resistant to EGFR-TKIs still demonstrated a better response to ani-PD-1/PD-L1 agents. The effect of EGFR-TKIs in combination with ICIs is currently being investigated in a large number of clinical trials, most of which have shown no significantly enhance clinical activity and result in a limitation of further active investigation due to the unexpectedly high incidence of irAEs. The inspiring results from the recent IMpower 150 trial made the chemoimmunotherapy regimen a possible therapeutic alternative for EGFR-TKI-resistant patients. At present, the drug resistance of EGFR-TKIs is being studied in depth. It is particularly necessary to explore the resistance mechanism to targeted therapy, additional potential targets and the development of effective biomarkers to screen the advantaged populations receiving immunotherapy for the realization of precision therapy in the future.

    In this review, we analyzed preclinical and clinical studies and concluded the following highly selective populations may benefit from immunotherapy: high expression of PD-L1 and TMB; EGFR L858R, G719X, EX20 insertion mutation and T790M negative; primary resistance to EGFR-TKIs; a long-term treatment duration of EGFR-TKIs and CD73 overexpression after resistance to EGFR-TKIs. Therefore, more research is required to provide the best-individualized treatment for EGFR-TKI-resistant patients. Further experiments confirm the synergistic anti-tumor effect of the combinational strategy of EGFR-TKIs and ICIs. This review summarized the four immunotherapy combinations for patients with NSCLC who fail EGFR-TKIs, mainly focused on immunotherapy-based combination therapy: immunotherapy in combination with EGFR-TKIs; immunotherapy in combination with chemotherapy; immunotherapy in combination with antiangiogenic agents and immunotherapy in combination with CTLA-4 inhibitors (Figure 2). Most of the clinical data are preliminary and immature, and more investigations and additional long-term follow-ups are required. Dramatic success from the ABCP regimen makes it a potentially promising therapy for those who fail EGFR-TKIs. It is hypothesized that a combination of multiple anti-tumor drugs will be employed to overcome EGFR-TKI resistance in the future. Better clinical outcomes with manageable adverse reactions may be achieved in the ongoing combinatorial clinical trials.

    Figure 2. Treatment strategies for patients with non-small-cell lung cancer with EGFR-tyrosine kinase inhibitor resistance.

    EMT: Epithelial-mesenchymal transition; ICI: Immune checkpoint inhibitor; NSCLC: Non-small-cell lung cancer; TKI: Tyrosine kinase inhibitor.

    Executive summary

    Background

    • Drug resistance to EGFR-tyrosine kinase inhibitors (TKIs) remains a challenge for both patients and clinicians, with no clear consensus on treatment.

    • The dynamic relationship between EGFR mutation status and PD-1 expression pre- and post-EGFR-TKI treatment in patients with EGFR mutations has previously been reported.

    • Preliminary results from serval clinical trials have shown favorable survival benefits for patients with non-small-cell lung cancer (NSCLC) resistant to EGFR-TKIs.

    Immunotherapy monotherapy in EGFR-TKI-resistant NSCLC

    • The limited efficacy of immunotherapy in EGFR-positive NSCLC has been described by early-phase trials.

    • EGFR mutated individuals usually establish an immunosuppressive noninflammatory tumor microenvironment, leading to resistance to immunotherapy.

    • High levels of PD-L1 expression and tumor mutational burden have been considered promising predictive biomarkers of immunological therapy efficacy in recent studies.

    EGFR-TKIs in combination with immunotherapy

    • EGFR-TKIs in combination with ICIs from a large number of clinical trials showed no significant enhanced clinical activity due to an unexpectedly high incidence of immune-related adverse events.

    • The optimal sequence, time interval and dosage of administration remain to be explored.

    Chemotherapy in combination with immunotherapy

    • Chemotherapy in tandem with immunotherapy may improve the anti-tumor response.

    • The atezolizumab plus bevacizumab plus carboplatin plus paclitaxel regimen is an innovative therapeutic option for post-line treatment of failure on EGFR-TKIs.

    Antiangiogenic agents in combination with immunotherapy

    • Preclinical studies corroborate the complementary therapeutic effects of this combination of antiangiogenic therapy and immunotherapy.

    • The combination of antiangiogenic therapy and immunotherapy displays promising efficacy in EGFR-TKI-resistant NSCLC individuals, particularly in those with high VEGF levels.

    CTLA-4 inhibitors in combination with immunotherapy

    • The combination of CTLA-4 and PD-1/PD-L1 inhibitors enhanced the anti-tumor activity in EGFR-TKI-resistant NSCLC, especially for those with higher tumor mutational burden and PD-L1expression.

    Financial & competing interests disclosure

    This study was supported by the National Natural Science Youth Foundation of China (82072561), the National Natural Science Youth Foundation of China (81501971), project funded by China Postdoctoral Science Foundation (2018M630603), the Natural Science Youth Foundation of Jiangsu Province (BK20150252), the Human Resource Summit Grant of Jiangsu Province (WSW-142) and the Youth Medical Professionals Foundation of Jiangsu Province (QNRC2016279). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J. Clin. 70(1), 7–30 (2020).
    • 2. Lara MS, Brunson A, Wun T et al. Predictors of survival for younger patients less than 50 years of age with non-small cell lung cancer (NSCLC): a California Cancer Registry analysis. Lung Cancer 85(2), 264–269 (2014).
    • 3. Edwards BK, Noone AM, Mariotto AB et al. Annual report to the nation on the status of cancer, 1975–2010, featuring prevalence of comorbidity and impact on survival among persons with lung, colorectal, breast, or prostate cancer. Cancer 120(9), 1290–1314 (2014).
    • 4. Hanna N, Johnson D, Temin S et al. Systemic therapy for stage IV non-small-cell lung cancer: American Society of Clinical Oncology Clinical Practice Guideline update. J. Clin. Oncol. 35(30), 3484–3515 (2017).
    • 5. Ettinger DS, Wood DE, Akerley W et al. NCCN guidelines insights: non-small cell lung cancer, version 4.2016. J. Natl Compr. Cancer Netw. 14(3), 255–264 (2016).
    • 6. Wu YL, Zhou C, Liam CK et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann. Oncol. 26(9), 1883–1889 (2015).
    • 7. Minari R, Bordi P, Tiseo M. Third-generation epidermal growth factor receptor-tyrosine kinase inhibitors in T790M-positive non-small cell lung cancer: review on emerged mechanisms of resistance. Transl. Lung Cancer Res. 5(6), 695–708 (2016).
    • 8. Huang L, Huang H, Zhou XP et al. Osimertinib or EGFR-TKIs/chemotherapy in patients with EGFR-mutated advanced nonsmall cell lung cancer: a meta-analysis. Medicine (Baltimore) 98(43), e17705 (2019).
    • 9. Pai-Scherf L, Blumenthal GM, Li H et al. FDA approval summary: pembrolizumab for treatment of metastatic non-small cell lung cancer: first-line therapy and beyond. Oncologist 22(11), 1392–1399 (2017).
    • 10. Horn L, Spigel DR, Vokes EE 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. 35(35), 3924–3933 (2017).
    • 11. Reck M. Pembrolizumab as first-line therapy for metastatic non-small-cell lung cancer. Immunotherapy 10(2), 93–105 (2018).
    • 12. Cyriac G, Gandhi L. Emerging biomarkers for immune checkpoint inhibition in lung cancer. Semin. Cancer Biol. 52(Pt 2), 269–277 (2018).
    • 13. Hsu KH, Huang YH, Tseng JS et al. High PD-L1 expression correlates with primary resistance to EGFR-TKIs in treatment-naive advanced EGFR-mutant lung adenocarcinoma patients. Lung Cancer 127, 37–43 (2019).
    • 14. Yang CY, Liao WY, Ho CC et al. Association between programmed death-ligand 1 expression, immune microenvironments, and clinical outcomes in epidermal growth factor receptor mutant lung adenocarcinoma patients treated with tyrosine kinase inhibitors. Eur. J. Cancer 124, 110–122 (2020). •• This article analyzed the relationship between PD-L1 and tumor microenvironment in detail and combined clinical trials to make the results convincing and credible.
    • 15. Haratani K, Hayashi H, Tanaka T et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann. Oncol. 28(7), 1532–1539 (2017).
    • 16. Isomoto K, Haratani K, Hayashi H et al. Impact of EGFR-TKI treatment on the tumor immune microenvironment in EGFR mutation-positive non-small cell lung cancer. Clin. Cancer Res. 26(8), 2037–2046 (2020).
    • 17. Okita R, Maeda A, Shimizu K, Nojima Y, Saisho S, Nakata M. PD-L1 overexpression is partially regulated by EGFR/HER2 signaling and associated with poor prognosis in patients with non-small-cell lung cancer. Cancer Immunol. Immunother. 66(7), 865–876 (2017).
    • 18. Tang Y, Fang W, Zhang Y et al. The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget 6(16), 14209–14219 (2015).
    • 19. Lin C, Chen X, Li M et al. Programmed death-ligand 1 expression predicts tyrosine kinase inhibitor response and better prognosis in a cohort of patients with epidermal growth factor receptor mutation-positive lung adenocarcinoma. Clin. Lung Cancer 16(5), e25–35 (2015).
    • 20. Li J, Chen Y, Shi X et al. A systematic and genome-wide correlation meta-analysis of PD-L1 expression and targetable NSCLC driver genes. J. Thorac. Dis. 9(8), 2560–2571 (2017). •• This meta-analysis systematically analyzed the relationship between PD-L1 and driver genes making the results clear and enabling people to have a clearer understanding of the relationship between them.
    • 21. Dong ZY, Zhang JT, Liu SY et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 6(11), e1356145 (2017).
    • 22. Hata A, Katakami N, Nanjo S et al. Programmed death-ligand 1 expression according to epidermal growth factor receptor mutation status in pretreated non-small cell lung cancer. Oncotarget 8(69), 113807–113816 (2017).
    • 23. Suda K, Rozeboom L, Furugaki K et al. Increased EGFR phosphorylation correlates with higher programmed death ligand-1 expression: analysis of TKI-resistant lung cancer cell lines. Biomed. Res. Int. 2017, 7694202 (2017).
    • 24. Hirsch FR, Mcelhinny A, Stanforth D et al. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project. J. Thorac. Oncol. 12(2), 208–222 (2017).
    • 25. Jia Y, Li X, Jiang T et al. EGFR-targeted therapy alters the tumor microenvironment in EGFR-driven lung tumors: implications for combination therapies. Int. J. Cancer 145(5), 1432–1444 (2019).
    • 26. Akbay EA, Koyama S, Carretero J et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 3(12), 1355–1363 (2013).
    • 27. Lastwika KJ, Wilson W 3rd, Li QK et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 76(2), 227–238 (2016).
    • 28. Abdelhamed S, Ogura K, Yokoyama S, Saiki I, Hayakawa Y. AKT-STAT3 pathway as a downstream target of EGFR signaling to regulate PD-L1 expression on NSCLC cells. J. Cancer 7(12), 1579–1586 (2016).
    • 29. Meng F, Wang F, Wang L, Wong SC, Cho WC, Chan LW. MiR-30a-5p overexpression may overcome EGFR-inhibitor resistance through regulating PI3K/AKT signaling pathway in non-small cell lung cancer cell lines. Front. Genet. 7, 197 (2016).
    • 30. Chen N, Fang W, Zhan J et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J. Thorac. Oncol. 10(6), 910–923 (2015).
    • 31. Antonangeli F, Natalini A, Garassino MC, Sica A, Santoni A, Di Rosa F. Regulation of PD-L1 expression by NF-kappaB in cancer. Front. Immunol. 11, 584626 (2020).
    • 32. Guo R, Li Y, Wang Z et al. Hypoxia-inducible factor-1alpha and nuclear factor-kappaB play important roles in regulating programmed cell death ligand 1 expression by epidermal growth factor receptor mutants in non-small-cell lung cancer cells. Cancer Sci. 110(5), 1665–1675 (2019).
    • 33. Gao SP, Mark KG, Leslie K et al. Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. J. Clin. Invest. 117(12), 3846–3856 (2007).
    • 34. Kitamura H, Ohno Y, Toyoshima Y et al. Interleukin-6/STAT3 signaling as a promising target to improve the efficacy of cancer immunotherapy. Cancer Sci. 108(10), 1947–1952 (2017).
    • 35. Johnson DE, O'Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signaling axis in cancer. Nat. Rev. Clin. Oncol. 15(4), 234–248 (2018).
    • 36. Zhang N, Zeng Y, Du W et al. The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int. J. Oncol. 49(4), 1360–1368 (2016).
    • 37. Ota K, Azuma K, Kawahara A et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin. Cancer Res. 21(17), 4014–4021 (2015).
    • 38. Kumar S, Sharawat SK. Epigenetic regulators of programmed death-ligand 1 expression in human cancers. Transl. Res. 202, 129–145 (2018).
    • 39. Su S, Dong ZY, Xie Z et al. Strong programmed death-ligand 1 expression predicts poor response and de novo resistance to EGFR tyrosine kinase inhibitors among NSCLC patients with EGFR mutation. J. Thorac. Oncol. 13(11), 1668–1675 (2018).
    • 40. Lin A, Wei T, Meng H, Luo P, Zhang J. Role of the dynamic tumor microenvironment in controversies regarding immune checkpoint inhibitors for the treatment of non-small cell lung cancer (NSCLC) with EGFR mutations. Mol. Cancer 18(1), 139 (2019). •• This preclinical study fundamentally revealed the reasons for the poor efficacy of PD-L1 inhibitors in the EGFR-mutant population by analyzing the tumor microenvironment in this setting.
    • 41. Lin K, Cheng J, Yang T, Li Y, Zhu B. EGFR-TKI down-regulates PD-L1 in EGFR mutant NSCLC through inhibiting NF-kappaB. Biochem. Biophys. Res. Commun. 463(1–2), 95–101 (2015).
    • 42. Jiang XM, Xu YL, Huang MY et al. Osimertinib (AZD9291) decreases programmed death ligand-1 in EGFR-mutated non-small cell lung cancer cells. Acta Pharmacol. Sin. 38(11), 1512–1520 (2017).
    • 43. Gainor JF, Shaw AT, Sequist LV et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin. Cancer Res. 22(18), 4585–4593 (2016).
    • 44. Azuma K, Ota K, Kawahara A et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann. Oncol. 25(10), 1935–1940 (2014).
    • 45. Inomata M, Azechi K, Takata N et al. Association of tumor PD-L1 expression with the T790M mutation and progression-free survival in patients with EGFR-mutant non-small cell lung cancer receiving EGFR-TKI therapy. Diagnostics (Basel) 10(12), 1006 (2020).
    • 46. Kobayashi K, Seike M, Zou F et al. Prognostic significance of NSCLC and response to EGFR-TKIs of EGFR-mutated NSCLC based on PD-L1 expression. Anticancer Res. 38(2), 753–762 (2018).
    • 47. Lin PL, Wu TC, Wu DW, Wang L, Chen CY, Lee H. An increase in BAG-1 by PD-L1 confers resistance to tyrosine kinase inhibitor in non-small cell lung cancer via persistent activation of ERK signaling. Eur. J. Cancer 85, 95–105 (2017).
    • 48. Zhang Y, Zeng Y, Liu T et al. The canonical TGF-beta/Smad signaling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer. Respir. Res. 20(1), 164 (2019).
    • 49. Engelman JA, Janne PA. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin. Cancer Res. 14(10), 2895–2899 (2008). • This review introduced the mechanism of EGFR-tyrosine kinase inhibitor resistance in detail, which can help people to have a more intuitive understanding. These findings may lead to clinical trials using newly designed targeted therapies that can overcome these resistance mechanisms.
    • 50. Tung JN, Lin PL, Wang YC, Wu DW, Chen CY, Lee H. PD-L1 confers resistance to EGFR mutation-independent tyrosine kinase inhibitors in non-small cell lung cancer via upregulation of YAP1 expression. Oncotarget 9(4), 4637–4646 (2018).
    • 51. Lisberg A, Cummings A, Goldman JW et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naive patients with advanced NSCLC. J. Thorac. Oncol. 13(8), 1138–1145 (2018).
    • 52. Lee CK, Man J, Lord S et al. Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer – a meta-analysis. J. Thorac. Oncol. 12(2), 403–407 (2017).
    • 53. Borghaei H, Paz-Ares L, Horn L et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373(17), 1627–1639 (2015).
    • 54. Herbst RS, Baas P, Kim DW et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 387(10027), 1540–1550 (2016).
    • 55. Hayashi H, Sugawara S, Fukuda Y et al. A randomized phase II study comparing nivolumab (NIVO) with carboplatin-pemetrexed (CbPEM) for patients (pts) with EGFR mutation-positive non-small cell lung cancer (NSCLC) who acquire resistance to tyrosine kinase inhibitors (TKIs) not due to a secondary T790M mutation (WJOG8515L). J. Clin. Oncol. 39(Suppl. 15), 9037–9037 (2021).
    • 56. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene 27(45), 5904–5912 (2008).
    • 57. Li HY, Mcsharry M, Bullock B et al. The tumor microenvironment regulates sensitivity of murine lung tumors to PD-1/PD-L1 antibody blockade. Cancer Immunol. Res. 5(9), 767–777 (2017).
    • 58. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19(11), 1423–1437 (2013).
    • 59. Yoon BW, Chang B, Lee SH. High PD-L1 expression is associated with unfavorable clinical outcome in EGFR-mutated lung adenocarcinomas treated with targeted therapy. Onco. Targets Ther. 13, 8273–8285 (2020).
    • 60. Goodman AM, Kato S, Bazhenova L et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16(11), 2598–2608 (2017).
    • 61. Datar I, Sanmamed MF, Wang J et al. Expression analysis and significance of PD-1, LAG-3, and TIM-3 in human non-small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin. Cancer Res. 25(15), 4663–4673 (2019).
    • 62. Griesing S, Liao BC, Yang JC. CD73 is regulated by the EGFR-ERK signaling pathway in non-small cell lung cancer. Anticancer Res. 41(3), 1231–1242 (2021).
    • 63. Passarelli A, Aieta M, Sgambato A, Gridelli C. Targeting immunometabolism mediated by CD73 pathway in EGFR-mutated non-small cell lung cancer: a new hope for overcoming immune resistance. Front. Immunol. 11, 1479 (2020).
    • 64. Toki MI, Mani N, Smithy JW et al. Immune marker profiling and programmed death ligand 1 expression across NSCLC mutations. J. Thorac. Oncol. 13(12), 1884–1896 (2018).
    • 65. Mazzaschi G, Madeddu D, Falco A et al. Low PD-1 expression in cytotoxic CD8(+) tumor-infiltrating lymphocytes confers an immune-privileged tissue microenvironment in NSCLC with a prognostic and predictive value. Clin. Cancer Res. 24(2), 407–419 (2018).
    • 66. Garassino MC, Cho BC, Kim JH et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study. Lancet Oncol. 19(4), 521–536 (2018).
    • 67. Choudhury NJ, Schoenfeld AJ, Flynn J et al. Response to standard therapies and comprehensive genomic analysis for patients with lung adenocarcinoma with EGFR exon 20 insertions. Clin. Cancer Res. 27(10), 2920–2927 (2021).
    • 68. Hastings K, Yu HA, Wei W et al. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer. Ann. Oncol. 30(8), 1311–1320 (2019).
    • 69. Chen K, Pan G, Cheng G et al. Immune microenvironment features and efficacy of PD-1/PD-L1 blockade in non-small cell lung cancer patients with EGFR or HER2 exon 20 insertions. Thorac. Cancer 12(2), 218–226 (2021).
    • 70. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 12(4), 237–251 (2012).
    • 71. Ahn MJ, Sun JM, Lee SH, Ahn JS, Park K. EGFR TKI combination with immunotherapy in non-small cell lung cancer. Expert Opin. Drug Saf. 16(4), 465–469 (2017).
    • 72. Wang S, Zhang Y, Wang Y et al. Amphiregulin confers regulatory T cell suppressive function and tumor invasion via the EGFR/GSK-3beta/Foxp3 axis. J. Biol. Chem. 291(40), 21085–21095 (2016).
    • 73. Dominguez C, Tsang KY, Palena C. Short-term EGFR blockade enhances immune-mediated cytotoxicity of EGFR mutant lung cancer cells: rationale for combination therapies. Cell Death Dis. 7(9), e2380 (2016).
    • 74. He S, Yin T, Li D et al. Enhanced interaction between natural killer cells and lung cancer cells: involvement in gefitinib-mediated immunoregulation. J. Transl. Med. 11, 186 (2013).
    • 75. Muro K, Chung HC, Shankaran V et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 17(6), 717–726 (2016).
    • 76. Yang JC, Shepherd FA, Kim DW et al. Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M-positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J. Thorac. Oncol. 14(5), 933–939 (2019).
    • 77. Wan N, Ji B, Li J et al. A pooled meta-analysis of PD-1/L1 inhibitors incorporation therapy for advanced non-small cell lung cancer. Onco. Targets Ther. 12, 4955–4973 (2019).
    • 78. Spigel DR, Reynolds C, Waterhouse D et al. Phase 1/2 study of the safety and tolerability of nivolumab plus crizotinib for the first-line treatment of anaplastic lymphoma kinase translocation – positive advanced non-small cell lung cancer (CheckMate 370). J. Thorac. Oncol. 13(5), 682–688 (2018).
    • 79. Gettinger S, Hellmann MD, Chow LQM et al. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J. Thorac. Oncol. 13(9), 1363–1372 (2018).
    • 80. Oxnard GR, Yang JC, Yu H et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 31(4), 507–516 (2020).
    • 81. Schoenfeld AJ, Arbour KC, Rizvi H et al. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann. Oncol. 30(5), 839–844 (2019).
    • 82. Gandhi L, Rodriguez-Abreu D, Gadgeel S et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N. Engl. J. Med. 378(22), 2078–2092 (2018).
    • 83. Soo RA, Lim SM, Syn NL et al. Immune checkpoint inhibitors in epidermal growth factor receptor mutant non-small cell lung cancer: current controversies and future directions. Lung Cancer 115, 12–20 (2018). • This review showed a greater understanding of the tumor microenvironment in the EGFR-tyrosine kinase inhibitor-resistant group and pointed out the feasibility and limitations of immune checkpoint inhibitors in EGFR-mutant non-small-cell lung cancer, bringing more profound thinking.
    • 84. Pacheco JM, Camidge DR, Doebele RC, Schenk E. A changing of the guard: immune checkpoint inhibitors with and without chemotherapy as first line treatment for metastatic non-small cell lung cancer. Front. Oncol. 9, 195 (2019).
    • 85. Saxena P, Singh PK, Malik PS, Singh N. Immunotherapy alone or in combination with chemotherapy as first-line treatment of non-small cell lung cancer. Curr. Treat. Options Oncol. 21(8), 69 (2020).
    • 86. Zhou Y, Chen C, Zhang X et al. Immune-checkpoint inhibitor plus chemotherapy versus conventional chemotherapy for first-line treatment in advanced non-small cell lung carcinoma: a systematic review and meta-analysis. J. Immunother. Cancer 6(1), 155 (2018).
    • 87. Wang Y, Ma X, Wei Y, Ma D, Gong P. Effect of platinum-based chemotherapy on EGFR gene mutation status in lung adenocarcinoma. Medicine (Baltimore) 97(4), e9602 (2018).
    • 88. Peng J, Hamanishi J, Matsumura N et al. Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-kappaB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res. 75(23), 5034–5045 (2015).
    • 89. Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Diff. 21(1), 15–25 (2014). •• This review provided an extensive overview of the multiple immune-based mechanisms exploited by the most commonly employed cytotoxic drugs, with the final aim of identifying prerequisites for optimal combination with immunotherapy strategies for the development of more effective treatments against cancer.
    • 90. Menard C, Martin F, Apetoh L, Bouyer F, Ghiringhelli F. Cancer chemotherapy: not only a direct cytotoxic effect, but also an adjuvant for antitumor immunity. Cancer Immunol. Immunother. 57(11), 1579–1587 (2008).
    • 91. Apetoh L, Ghiringhelli F, Tesniere A et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13(9), 1050–1059 (2007).
    • 92. Leonetti A, Wever B, Mazzaschi G et al. Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non-small cell lung cancer. Drug Resist. Update 46, 100644 (2019).
    • 93. Champiat S, Ileana E, Giaccone G et al. Incorporating immune-checkpoint inhibitors into systemic therapy of NSCLC. J. Thorac. Oncol. 9(2), 144–153 (2014).
    • 94. Borghaei H, Langer CJ, Paz-Ares L et al. Pembrolizumab plus chemotherapy versus chemotherapy alone in patients with advanced non-small cell lung cancer without tumor PD-L1 expression: a pooled analysis of 3 randomized controlled trials. Cancer 126(22), 4867–4877 (2020).
    • 95. Schvartsman G, Peng SA, Bis G et al. Response rates to single-agent chemotherapy after exposure to immune checkpoint inhibitors in advanced non-small cell lung cancer. Lung Cancer 112, 90–95 (2017).
    • 96. Zhang J, Zhou CC, Zhao Y et al. MA11.06 A PII study of toripalimab, a PD-1 mAb, in combination with chemotherapy in EGFR+ advanced NSCLC patients failed to prior EGFR TKI therapies. J. Thorac. Oncol. 14 (10), S292 (2019).
    • 97. Reck M, Mok TSK, Nishio M et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir. Med. 7(5), 387–401 (2019). •• This randomized phase III clinical trial was the first to show an overall survival benefit of immunotherapy-based combination strategies in patients with EGFR-sensitive mutated non-small-cell lung cancer, which suggested that the atezolizumab plus bevacizumab plus carboplatin plus paclitaxel regimen breaks the poor effect of immunotherapy in EGFR-mutated non-small-cell lung cancer, and will be a new option for post-line treatment after failure of EGFR-tyrosine kinase inhibitor therapy.
    • 98. Aredo JV, Mambetsariev I, Hellyer JA et al. Durvalumab for stage III EGFR-mutated NSCLC after definitive chemoradiotherapy. J. Thorac. Oncol. 16(6), 1030–1041 (2021).
    • 99. Socinski MA, Jotte RM, Cappuzzo F et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378(24), 2288–2301 (2018).
    • 100. Siveen KS, Prabhu K, Krishnankutty R et al. Vascular endothelial growth factor (VEGF) signaling in tumour vascularization: potential and challenges. Curr. Vasc. Pharmacol. 15(4), 339–351 (2017).
    • 101. Hendry SA, Farnsworth RH, Solomon B, Achen MG, Stacker SA, Fox SB. The role of the tumor vasculature in the host immune response: implications for therapeutic strategies targeting the tumor microenvironment. Front. Immunol. 7, 621 (2016).
    • 102. Lapeyre-Prost A, Terme M, Pernot S et al. Immunomodulatory activity of VEGF in cancer. Int. Rev. Cell Mol. Biol. 330, 295–342 (2017).
    • 103. Liang H, Wang M. Prospect of immunotherapy combined with anti-angiogenic agents in patients with advanced non-small cell lung cancer. Cancer Manag. Res. 11, 7707–7719 (2019).
    • 104. Nakamura K, Smyth MJ. Targeting cancer-related inflammation in the era of immunotherapy. Immunol. Cell Biol. 95(4), 325–332 (2017).
    • 105. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 15(5), 325–340 (2018).
    • 106. Zhao S, Ren S, Jiang T et al. Low-dose apatinib optimizes tumor microenvironment and potentiates antitumor effect of PD-1/PD-L1 blockade in lung cancer. Cancer Immunol. Res. 7(4), 630–643 (2019).
    • 107. Wu J, Zhao X, Sun Q et al. Synergic effect of PD-1 blockade and endostar on the PI3K/AKT/mTOR-mediated autophagy and angiogenesis in Lewis lung carcinoma mouse model. Biomed. Pharmacother. 125, 109746 (2020).
    • 108. Xiong Q, Qin B, Xin L et al. Real-world efficacy and safety of anlotinib with and without immunotherapy in advanced non-small cell lung cancer. Front. Oncol. 11, 659380 (2021).
    • 109. Zhao M, Han T. Camrelizumab monotherapy or in combination with apatinib for PD-L1-positive advanced pulmonary sarcomatoid carcinoma: a multicenter, open-label, single-arm, phase II study. J. Clin. Oncol. 39(Suppl. 15), TPS9130 (2021).
    • 110. Qiu L, Zhao X, Shi W et al. Real-world treatment efficacy of anti-programmed death-1 combined with anti-angiogenesis therapy in non-small cell lung cancer patients. Medicine (Baltimore) 99(24), e20545 (2020).
    • 111. Kibirova A, Mattes MD, Smolkin M, Ma PC. The journey of an EGFR-mutant lung adenocarcinoma through erlotinib, osimertinib and ABCP immunotherapy regimens: sensitivity and resistance. Case Rep. Oncol. 12(3), 765–776 (2019).
    • 112. Zhang Y, Zhao M, Cao S, Zhang X, Du Y. Unexpected favorable outcome to sintilimab plus bevacizumab in an EGFR-mutated non-small cell lung cancer patient: a case report. Thorac. Cancer 11(9), 2717–2722 (2020).
    • 113. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008).
    • 114. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl Acad. Sci. USA 99(19), 12293–12297 (2002).
    • 115. Ni L, Dong C. New B7 family checkpoints in human cancers. Mol. Cancer Ther. 16(7), 1203–1211 (2017).
    • 116. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 224, 166–182 (2008).
    • 117. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 39(1), 98–106 (2016).
    • 118. Gide TN, Quek C, Menzies AM et al. Distinct immune cell populations define response to anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell 35(2), 238–255 e236 (2019).
    • 119. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA 107(9), 4275–4280 (2010).
    • 120. Santarpia M, Giovannetti E, Rolfo C et al. Recent developments in the use of immunotherapy in non-small cell lung cancer. Expert Rev. Respir. Med. 10(7), 781–798 (2016).
    • 121. Ott PA, Hodi FS, Kaufman HL, Wigginton JM, Wolchok JD. Combination immunotherapy: a road map. J. Immunother. Cancer 5, 16 (2017).
    • 122. Hodi FS, O'Day SJ, Mcdermott DF et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363(8), 711–723 (2010).
    • 123. Hellmann MD, Rizvi NA, Goldman JW et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol. 18(1), 31–41 (2017).
    • 124. Ready N, Hellmann MD, Awad MM et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death-ligand 1 and tumor mutational burden as biomarkers. J. Clin. Oncol. 37(12), 992–1000 (2019).
    • 125. Gubens MA, Sequist LV, Stevenson JP et al. Pembrolizumab in combination with ipilimumab as second-line or later therapy for advanced non-small-cell lung cancer: KEYNOTE-021 cohorts D and H. Lung Cancer 130, 59–66 (2019).
    • 126. Rizvi NA, Cho BC, Reinmuth N et al. Durvalumab with or without tremelimumab vs standard chemotherapy in first-line treatment of metastatic non-small cell lung cancer: The MYSTIC phase 3 randomized clinical trial. JAMA Oncol. 6(5), 661–674 (2020).
    • 127. Reck M, Ciuleanu TE, Lee JS et al. First-line nivolumab plus ipilimumab versus chemotherapy in advanced NSCLC with 1% or greater tumor PD-L1 expression: patient-reported outcomes from CheckMate 227 Part 1. J. Thorac. Oncol. 16(4), 665–676 (2021).
    • 128. Planchard D, Barlesi F, Gomez-Roca C et al. Phase I, safety, tolerability and preliminary efficacy study of tremelimumab (Trem) in combination with gefitinib (Gef) in EGFR-mutant (EGFR-mut) NSCLC (GEFTREM). Ann. Oncol. 14(12 Suppl. 2), A51 (2016).
    • 129. Yang B, Long Y, Hu Y. Chemoimmunotherapy combination: potential option for metastatic NSCLC patients with EGFR mutation resistant to osimertinib. J. Clin. Oncol. 37(Suppl. 15), e20618–e20618 (2019).