Skip to main content
Chinese Medical Journal logoLink to Chinese Medical Journal
. 2021 Aug 2;134(16):1908–1919. doi: 10.1097/CM9.0000000000001610

Combination strategies of immunotherapy in non-small cell lung cancer: facts and challenges

Chu-Ling Li 1, Yong Song 1
Editor: Pei-Fang Wei1
PMCID: PMC8382384  PMID: 34343148

Abstract

Immunotherapy has dramatically altered the treatment of non-small cell lung cancer. Currently, the emergence of combination strategies in immunotherapy has brightened the prospects of improved clinical outcomes and manageable safety profiles in the first/second-line settings. However, sub-optimal response rates are still observed in several clinical trials. Hence, alternative combination models and candidate selection strategies need to be explored. Herein, we have critically reviewed and commented on the published data from several clinical trials, including combined immunotherapy and chemotherapy, anti-angiogenic agents, epidermal growth factor receptor/anaplastic lymphoma kinase tyrosine kinase inhibitors, radiotherapy, and other immune checkpoint inhibitors.

Keywords: Immunotherapy, Programmed death 1/programmed death ligand 1, Immune checkpoint inhibitors, Non-small cell lung cancer

Introduction

Lung cancer is the most commonly encountered cancer and is a leading cause of cancer-related deaths worldwide,[1] with approximately 80% of the cases being non-small cell lung cancer (NSCLC). In the past decade, immune checkpoint inhibitors (ICIs) such as programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors have dramatically altered the therapeutic strategy of NSCLC.[2] Based on previous clinical trials, pembrolizumab and nivolumab (PD-1 inhibitors) and atezolizumab (PD-L1 inhibitor) have been approved by the Food and Drug Administration (FDA) for the first/second-line treatment of metastatic NSCLC, while durvalumab (PD-L1 inhibitor) has been approved for stage III unresectable NSCLC.

PD-L1 is an immunosuppressive molecule expressed by tumor cells (TCs). PD-1/PD-L1 axis induces T-cell apoptosis and inhibits anti-tumor immunity in the tumor microenvironment (TME),[3,4] thereby accelerating the infiltration of cancer cells. Agents targeting PD-1/PD-L1 are designed to treat NSCLC by regulating anti-tumor immunity.

Despite the advances in immunotherapy, acquired resistance and limited efficacy are observed during single ICI therapy.[5] Recent studies have established that traditional therapies such as chemotherapy and/or targeted therapy alter the immunogenic patterns.[6] Therefore, efforts have been made to combine different strategies of ICIs with chemotherapy, anti-angiogenic therapy, targeted therapy, or therapy using agents targeting other immune checkpoints to enhance the treatment efficacy. This review has summarized the available data on combination therapy using ICIs, with an emphasis on candidate selection and adverse events [Table 1].

Table 1.

Several results from clinical trials evaluating the combination strategies of anti-PD-(L)1 agent.

Agents Study Line Subtype Treatment arms ORR (P) PFS (months) PFS HR (95% CI, P) OS (months) OS HR (95% CI, P)
Pembrolizumab KEYNOTE-021 First-line NSq Cohort G: Pembro + pemetrexed-carboplatin Pemetrexed-carboplatin 58% vs. 33% 24.5 vs. 9.9 0.54 (0.35–0.83) 34.5 vs. 21.1 0.71 (0.45–1.12)
KEYNOTE-189 First-line NSq Pembro + pemetrexed-cisplatin/carboplatin Pemetrexed-cisplatin/carboplatin 48.0% vs. 19.4% (<0.001) 9.0 vs. 4.9 0.48 (0.40–0.58, <0.001) 22.0 vs. 10.7 0.56 (0.45–0.70, <0.001)
KEYNOTE-407 First-line Sq Pembro + carboplatin-paclitaxel/nab-paclitaxel Carboplatin-paclitaxel/nab-paclitaxel 62.6% vs. 38.4% 8.0 vs. 5.1 0.57 (0.47–0.69, <0.001) 17.1 vs. 11.6 0.71 (0.58–0.88, <0.001)
PROLUNG Second-line NSq + Sq Pembro + docetaxel Docetaxel 42.5% vs. 15.8% (0.01) 9.5 vs. 3.9 0.24 (0.13–0.46, <0.001)
KEYNOTE-598 First-line NSq + Sq Pembro + ipilimumab Pembro + placebo 42.4% vs. 42.4% 8.2 vs. 8.4 1.06 (0.86–1.30, 0.72) 21.4 vs. 21.9 1.08 (0.85–1.37, 0.74)
KEYNOTE-799 First-line A: NSq + Sq B: NSq A: Pembro + pacilitaxel-carboplatin + radiotherapy B: Pembro + pemetrexed-cisplatin + radiotherapy 69.6% 70.5%
Nivolumab CheckMate 227 First-line NSq + Sq Nivo + ipilimumab Platinum-based chemotherapy 36.4% vs. 30.2% 17.1 vs. 14.9 0.79 (0.67–0.93, 0.033)
CheckMate 9LA First-line NSq + Sq Nivo + ipilimumab + chemotherapy Chemotherapy 38.2% vs. 24.9% 6.7 vs. 5.0 0.68 (0.57–0.82) 15.6 vs. 10.9 0.66 (0.55–0.80)
Atezolizumab IMpower130 First-line NSq Atezo + carboplatin-nab-paclitaxel Carboplatin-nab-paclitaxel 49% vs. 32% 7.0 vs. 5.5 0.64 (0.54–0.77, <0.0001) 18.6 vs. 13.9 0.79 (0.64–0.98, 0.033)
IMpower131 First-line Sq Atezo + carboplatin-nab-paclitaxel Carboplatin-nab-paclitaxel 49.7% vs. 41.0% 6.3 vs. 5.6 0.71 (0.60–0.85, 0.0001) 14.2 vs. 13.5 0.88 (0.73–1.05, 0.1631)
IMpower132 First-line NSq Atezo + pemetrexed-cisplatin/carboplatin Pemetrexed-cisplatin/carboplatin 47% vs. 32% 7.6 vs. 5.2 0.60 (0.49–0.72, <0.0001) 17.5 vs. 13.6 0.86 (0.71–1.06, 0.1546)
IMpower150 First-line NSq Atezo + carboplatin-paclitaxel Bevacizumab-carboplatin-paclitaxel 63.5% vs. 48.0% 8.3 vs. 6.8 0.62 (0.52–0.74, <0.001) 19.2 vs. 14.7 0.78 (0.64–0.96, 0.02)
Durvalumab MYSTIC First-line NSq + Sq Durva + tremelimumab Platinum-based chemotherapy 34.4% vs. 37.7% 3.9 vs. 5.4 1.05 (0.72–1.53, 0.71) 11.9 vs. 12.9 0.85 (0.61–1.17, 0.20)
PACIFIC First-line NSq + Sq Durva + chemoradiotherapy Placebo + chemoradiotherapy 28.4% vs. 16.0% (<0.001) 16.8 vs. 5.6 0.52 (0.42–0.65, <0.001) Not reach vs. 29.1 0.69 (0.55–0.86, 0.20)

PD-L1 TPS ≥1%. CI: Confidence interval; HR: Hazard ratio; NSq: Non-squamous; ORR: Objective response rate; OS: Overall survival; PD-(L)1: Programmed death (ligand) 1; PFS: Progression free survival; Sq: Squamous; TPS: Tumor proportion score.

Immunotherapy Combined with Other Treatments

Efficacy of ICIs when combined with chemotherapy

Accumulating evidence has indicated that chemotherapy augments the immunologic effects of ICIs via various molecular mechanisms [Figure 1].[7] Chemotherapy can induce immunogenic cell death, which releases danger-associated molecular patterns and tumor-associated antigens to recruit antigen-presenting cells.[8] Thus, tumor infiltrating CD8+ T-cells are cross-primed, whereas myeloid derived suppressor cells and regulatory T cells are downregulated.[911] This complex regulation of immune cells (ICs) is accompanied by an increase in PD-L1 expression or tumor antigen presentation.[12] Based on these findings, several clinical trials have been conducted to investigate the efficacy and safety of ICIs when combined with platinum-based chemotherapy.

Figure 1.

Figure 1

Effect of chemotherapy on the modulation of tumor microenvironment. DAMPs: Danger-associated molecular patterns; MHC: Major histocompatibility complex; PD-1: Programmed death 1; PD-L1: Programmed death ligand 1; TAA: Tumor-associated antigen.

KEYNOTE-021,[13] a multi-center, open-label, randomized clinical trial was designed to compare the efficacies of pembrolizumab plus carboplatin-pemetrexed, and carboplatin-pemetrexed. The findings revealed that pembrolizumab with carboplatin-pemetrexed was the most effective combination. After a median follow-up period of 49.4 months, cohort G demonstrated significant improvement with pembrolizumab plus carboplatin-pemetrexed vs. carboplatin-pemetrexed in objective response rate (ORR) (58% vs. 33%) as well as progression-free survival (PFS) (24.5 vs. 9.9 months; hazard ratio [HR]: 0.54; 95% confidence interval [CI]: 0.35–0.83). However, the overall survival (OS) of the two groups was comparable.[14] Based on this trial, the FDA accelerated the approval of pembrolizumab in combination with carboplatin plus pemetrexed for the first-line treatment of non-squamous NSCLC. KEYNOTE-189[15] validated this finding in previously untreated metastatic non-squamous NSCLC lacking epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) mutations. Crossover to pembrolizumab was allowed at progression. The latest updated analysis of a median follow-up of 23.1 months revealed that the OS was significantly prolonged in the pembrolizumab-combination group when compared with the placebo-combination group at 22.0 vs. that at 10.7 months, respectively (HR: 0.56; 95% CI: 0.45–0.70).[16] Meanwhile, both studies performed exploratory analysis based on the level of PD-L1 expression, which indicated that pembrolizumab combined with chemotherapy exhibited better performance regardless of PD-L1 expression. Similar OS improvement was observed for squamous NSCLC patients treated with pembrolizumab plus chemotherapy over placebo plus chemotherapy (17.1 vs. 11.6 months) in KEYNOTE-407.[17]

Atezolizumab is another PD-L1 monoclonal antibody offering substantial clinical benefit when combined with chemotherapy. The clinical trial IMpower130[18] was designed to evaluate the efficacy and safety of atezolizumab plus chemotherapy and compare it with chemotherapy alone as a first-line therapy for metastatic non-squamous NSCLC. The latest results suggested that combining atezolizumab with chemotherapy enabled significant improvement in both OS (18.6 vs. 13.9 months) and PFS (7.0 vs. 5.5 months), except in the liver metastases subgroup and EGFR/ALK mutation subgroup. Furthermore, 45% of the patients in the combination group suffered from immune-related adverse events (irAEs), most of which were grade 1 or 2. Two other trials, IMpower131[19] and IMpower132,[20] highlighted the superiority of the combination therapy, asserting that atezolizumab combined with carboplatin and nab-paclitaxel (A + CnP) or carboplatin/cisplatin and pemetrexed (APP) significantly improved the PFS but not OS in first-line squamous and non-squamous NSCLC settings when compared with platinum-based chemotherapy.

Immunotherapy combined with chemotherapy has been immensely successful in the first-line setting. Some combinations provided benefits for patients with disease progression after traditional platinum-based chemotherapy. A phase II trial, PROLUNG,[21] demonstrated that, when compared to docetaxel alone, pembrolizumab combined with docetaxel improved the ORR (42.5% vs. 15.8%) and PFS (9.5 vs. 3.9 months) in NSCLC patients who had previously undergone chemotherapy, including those with EGFR mutations. KEYNOTE-789 is an ongoing clinical study comparing the efficacies of pembrolizumab plus platinum-based chemotherapy and platinum-based chemotherapy alone after the progression of first-line targeted tyrosine kinase inhibitors (TKIs) in advanced NSCLC patients with EGFR mutation. The data have not yet been officially published, and we look forward to the final performance.

Efficacy of ICI combinations

CheckMate 012[22] was a phase I study designed to assess the efficacy and safety of nivolumab plus ipilimumab for NSCLC. Patients receiving different doses of nivolumab plus ipilimumab as first-line therapy were compared in all the PD-L1 expression cohorts. The results implied that the performance of ICI combination therapy was superior, particularly in the cohorts with positive PD-L1 expression. A retrospective analysis of CheckMate 012 uncovered that a higher tumor mutation burden (TMB) assessed by whole exome sequencing (WES), which was independent of PD-L1 levels, was correlated with superior ORR and PFS in the immunotherapy-combination arm.[23] In another phase III trial, CheckMate 227,[24] stage IV or recurrent NSCLC patients were randomly assigned in a 1:1:1 ratio. Patients with PD-L1 ≥1% received nivolumab plus ipilimumab, nivolumab, or chemotherapy alone, whereas those with PD-L1 <1% received nivolumab plus ipilimumab, nivolumab plus chemotherapy, or chemotherapy alone. The median OS was observed 17.1 months with nivolumab plus ipilimumab and 13.9 months with chemotherapy alone. A 3-year update revealed that patients with PD-L1 ≥1% derived survival benefit from nivolumab plus ipilimumab vs. chemotherapy (HR: 0.79; 95% CI: 0.67–0.93); this improvement in health-related quality of life was also observed in patients treated with nivolumab plus ipilimumab.[25,26] CheckMate 9LA further investigated whether the addition of a limited course (two cycles) of chemotherapy to this combination would enhance the benefit. Recently, it revealed prolonged OS in the experimental group than that in the control group (15.6 vs. 10.9 months).[27]

On the other hand, KEYNOTE-598 compared the efficacy of pembrolizumab plus ipilimumab vs. that of pembrolizumab alone for NSCLC with PD-L1 tumor proportion score ≥50%. As a result, the addition of ipilimumab to pembrolizumab did not improve the overall efficacy with the median PFS of 8.2 and 8.4 months for pembrolizumab-ipilimumab and pembrolizumab-placebo treatments, respectively. In addition, the combination therapy was associated with greater toxicity than monotherapy.[28] Another three trials (MYSTIC, NEPTUNE, and ARTIC) have evaluated the combination of durvalumab and tremelimumab (either completed or ongoing). Recently, the open-label, phase III randomized MYSTIC[29] trial compared durvalumab with or without tremelimumab and chemotherapy as the first-line therapies for metastatic NSCLC patients (except in the case of EGFR or ALK alterations). A total of 1118 patients were randomized, with OS and PFS being the primary endpoints. It was disappointing that this study did not meet the primary endpoints of prolonged OS or PFS with the use of immunotherapy combinations. Even though the results are yet to be officially published, a recent press release from AstraZeneca revealed that this combination strategy did not meet its endpoint and that it failed to show improved survival in NEPTUNE[30] as well. Another trial, ARTIC,[31] also failed to improve the clinical benefit in the combination arm.

Past studies with ICI combinations mentioned earlier provided completely different bipolar outcomes, which has raised some questions, for example, whether it is “less is more” or “the more the better.” The combination of nivolumab and ipilimumab suggested advantages over monotherapy at multiple endpoints, while the combination of pembrolizumab and ipilimumab or durvalumab and tremelimumab indicated more serious AEs. As a result, some questions are raised, for example, whether there are any differences between different PD-(L)1 monoclonal antibodies; how can one select from different options for the advantaged population; and how can further extend research in this field.

Efficacy of ICIs when combined with anti-angiogenics

Anti-angiogenic agents block the vascular endothelial growth factor (VEGF), thereby exerting immunomodulatory effects, including promotion of the infiltration of T cells in the tumor, acceleration of dendritic cell maturation, and prevention of the immunosuppressive TME.[32] Current studies focusing on the cross-talk between the tumor and its microenvironment have provided opportunities for combining ICIs and anti-angiogenics.

The clinical trial IMpower150[33] demonstrated improved PFS and OS with the use of atezolizumab plus bevacizumab plus carboplatin plus paclitaxel (ABCP) when compared with bevacizumab plus carboplatin plus paclitaxel (BCP) in chemotherapy-naïve patients independent of the PD-L1 expression. Similarly, the LEAP-006[34] trial evaluated the first-line lenvatinib (multiple-receptor TKI of VEGFR 1–3, fibroblast growth factor receptor 1–4, platelet-derived growth factor receptor α, c-kit, and rearranged during transfection proto-oncogene [RET]) with pembrolizumab plus chemotherapy for metastatic non-squamous NSCLC. This trial is ongoing and expected to enroll approximately 714 patients across countries. WJOG11218L/APPLE[35] is another ongoing study that aims to explore the efficacy of atezolizumab with platinum-pemetrexed and with/without bevacizumab for non-squamous NSCLC.

Notably, the ABCP regimen in IMpower150 also provides clinical benefits for patients with EGFR mutations. These data together suggest that an understanding of the PD-1/PD-L1 axis and its associated regulatory proteins can be leveraged to improve the treatment efficacy, including for those who do not respond well to ICIs monotherapy. In the future, for patients harboring sensitive EGFR mutations, we could explore the options of combination therapy and determine suitable treatment options.

Efficacy of ICIs combined with TKIs

In the treatment-naïve NSCLC patients with EGFR or ALK alterations, EGFR/ALK TKIs remain the standard treatment. Most prospective clinical trials have also excluded patients with EGFR mutations or ALK rearrangements. Therefore, only limited randomized data were available in this regard.

KEYNOTE-021[36] tested the efficacy of pembrolizumab in combination with TKIs as the first-line therapy for advanced NSCLC patients containing sensitizing EGFR mutation. The study unearthed that the combination treatment group exhibited longer median PFS than the group receiving first generation EGFR-TKIs or osimertinib (19.5 vs. 11.0 vs. 19.2 months). The CheckMate 012[37] trial involved the EGFR-mutant subgroup treated with nivolumab and erlotinib. A total of 21 NSCLC patients, 20 of whom were pretreated with TKIs, were enrolled. According to the interim data, the ORR was 15% and five had treatment-related grade 3 toxicities. Similar safety data have been reported for the durvalumab plus gefitinib (grade 3–4 toxicities, 20%)[38] and atezolizumab plus erlotinib (grade 3–4 toxicities, 39%)[39] cohorts in TKI-naïve patients.

The phase 1b TATTON study evaluating the combination of durvalumab with osimertinib in patients with EGFR mutation was terminated because of the occurrence of interstitial pneumonia (38%). Owing to toxicity concerns, the phase 3 CAURAL trial comparing the combination of durvalumab and osimertinib with osimertinib alone in EGFR T790M positive NSCLC patients was also prematurely closed.

Another study, CheckMate 370, explored the efficacy and safety of nivolumab combined with crizotinib in NSCLC patients with ALK rearrangements. Severe hepatic toxicities were observed in the nivolumab plus crizotinib cohort (38%).[40] However, atezolizumab combined with alectinib only resulted in tolerable side effects related to the skin (18.9%).[41] Based on the outcomes of these clinical trials, it could be inferred that the combination of ICIs and TKIs is associated with a high incidence of toxicities. Overall, the efficacy and safety of the combined use of ICIs and EGFR/ALK-TKIs remain controversial.

Efficacy of ICIs combined with radiotherapy

Radiotherapy can kill cancer cells while simultaneously triggering pro-inflammatory mediators that release and increase ICs infiltration, thereby turning immunologically “cold” tumor into a “hot” one.[42] Based on this mechanism, radiotherapy, through its immuno-modulating effect, represents a promising combination alternative with ICIs.

PACIFIC[43] study compared durvalumab as a consolidation therapy with placebo in patients with unresectable stage III disease with non-progression after ≥2 cycles of platinum-based concurrent chemoradiation therapy (CCRT). Updated OS data (median follow-up duration: 33.3 months) demonstrate the long-term clinical benefit with durvalumab when compared with placebo (not reaching vs. 29.1 months; HR: 0.69; 95% CI: 0.55–0.86).[44] LUN 14-179 is another study that evaluated pembrolizumab as a consolidation therapy after CCRT in stage-III NSCLC patients. The primary endpoint, which is the time to metastatic disease or death, reached 30.7 months after the median follow-up of 32.2 months, which was significantly longer than the control period of 12.0 months.[45] Similar to the PACIFIC study and the LUN14-179 study, the KEYNOTE-799 demonstrated promising anti-tumor activity and tolerable toxicity of pembrolizumab plus CCRT in patients with unresectable stage-III NSCLC. This study was unique in bringing the time of immunotherapy forward from the maintenance to the synchronization treatment of CCRT.[46]

Presently, on the basis of these researches, the exploration of ICIs plus radiotherapy treatment in NSCLC continues to move forward. The efficacy and safety of this combined immunotherapy strategy are expected to be verified in more phase III studies.

Efficacy of ICIs when combined with other agents

Apart from combinatory therapies with chemotherapy, anti-angiogenics, other ICIs, TKIs or radiotherapy, emerging compounds which have been specifically designed to modulate the co-stimulatory/inhibitory receptors currently in combination with PD-(L)1 have been studied in combination therapy with ICIs. According to their targets, drugs have been classified into lymphoid inhibitor, lymphoid stimulators, and non-lymphoid inhibitors, non-lymphoid stimulators.[47]

T-cell immunoglobulin- and mucin-domain-containing molecule 3 (TIM-3) is a co-signaling molecule expressed by several ICs, which were verified to be associated with resistance to PD-1 inhibitors. Therefore, some clinical trials (NCT03099109, NCT03307785, NCT03311412, etc) are presently exploring TIM-3 inhibitor in combination with PD-(L)1 inhibitors for NSCLC.[48] Other lymphoid inhibitors including lymphocyte activation gene 3 protein inhibitor (NCT02460224, NCT02966548, NCT03849469, etc), T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain inhibitor (NCT02794571, NCT03628677, NCT03119428, etc),[49] and adenosine signal receptor blockade (A2AR, A2BR) (NCT04262856, NCT02403193, etc) were also tested for the safety and efficacy with PD-(L)1 inhibitor in NSCLC.

Indoleamine 2,3-dioxygenase (IDO) is a cytosolic enzyme. The upregulation of the IDO expression was observed after PD-L1 treatment, which indicated a candidate mechanism of resistance to ICIs.[50] IDO inhibitor belongs to non-lymphoid inhibitor. Several phase I clinical trials are investigating the efficacy and safety of IDO inhibitors, both in monotherapy and in combinatorial strategy in NSCLC (NCT02327078, NCT03085914, NCT02862457, NCT02178722, etc).[51] Clinical trials on combination therapies with lymphoid stimulators including OX40 agonists (NCT02410512, NCT03241173, etc) and glucocorticoid-induced tumor necrosis factor receptor-related gene agonists (NCT02740270), and non-lymphoid stimulators including anti-semaphorin 4D (SEMA4D) (NCT03268057), and NKTR-214 (NCT02983045, NCT03138889) are underway.

In addition to these immune modulators, the use of the combination of anti-PD-(L)1 therapy with other agents is also an attractive approach for NSCLC treatment. Chimeric antigen receptor (CAR)-T-cell therapy, as novel agents of immunotherapy, shows great promise for NSCLC. Currently, autologous CAR-T cells targeting PD-L1 and CD80/86 are being tested for the treatment of recurrent NSCLC in a phase I study (NCT03060343). Anti-PD-L1 CAR-T-cell therapy has also been used in metastatic PD-L1-positive NSCLC (NCT03330834).[52] Histone deacetylase inhibitors can enhance tumor immunogenicity through various mechanisms. A past study demonstrated that the combination of pembrolizumab plus vorinostat was associated with a considerably higher ORR than pembrolizumab monotherapy (NCT02638090).[53] JASPER (NCT03308942) is a phase-II study that has explored the combination of anti-PD-1 and niraparib (a poly adenosine diphosphate [ADP] ribose polymerase inhibitor) in metastatic NSCLC.[54] It showed that niraparib plus pembrolizumab induced durable response in NSCLC without new adverse events.

These studies represent the most promising therapies. Although these new therapies are still in the early phases of exploration, there is a need to undertake additional pre-clinical and clinical studies and be cautious in drawing conclusions.

Comments and Future Challenges

Patient selection

PD-L1

Although immunotherapy has been successful in the treatment of NSCLC, the response rate remains sub-optimal with ICIs. More than half of the unselected patients fail to benefit from immunotherapy.

To date, assessing PD-L1 expression by immunohistochemistry (IHC) is the only diagnostic test for ICIs that has been approved by the FDA. The expression of PD-L1 was determined by evaluating the percentage of TCs and ICs exhibiting a certain membrane expression intensity. In clinical trials, it was noted that PD-L1 expression is not an ideal biomarker for immunotherapy. Some patients with highly positive PD-L1 expression failed to benefit from ICIs, whereas about 10% of NSCLC patients with negative PD-L1 expression demonstrated reasonable response to ICIs.[5557] Similarly, NSCLC patients with PD-L1-negative tumors might benefit equally or more from a combination regimen,[16,25] but some patients with PD-L1 tumor proportion score (TPS)≥50% did not acquire improved efficacy.[58] This finding could be attributed to variable PD-L1 IHC assays (22C3, 28-8, SP142, and SP263 for pembrolizumab, nivolumab, atezolizumab, and durvalumab, respectively) and platforms (Dako and Ventana). The Blueprint project[59,60] was designed to examine the consistency of different IHC assays, and revealed that the 22C3, 28-8, and SP263 assays were highly aligned but not SP142. In addition, surgically resected specimens and biopsies showed inconsistent PD-L1 expression.[61]

The use of PD-L1 as a marker in immunotherapy has some challenges. For the clinical application of PD-L1 testing, there are strict requirements regarding the quality and source of specimens. Moreover, no antibody has so far been approved by the National Medical Products Administration for clinical testing and there is also a lack of standards for PD-L1 IHC specifications. Therefore, owing to interobserver difference, technological limitations, and tumor heterogeneity, a more appropriate diagnostic and reporting process is urgently needed for more extensive PD-L1 testing.

TMB and neoantigens

In recent years, TMB has been considered as another candidate biomarker since some somatic mutations generate tumor neoantigens that can be better recognized and captured by the immune system through protein translation. This view was supported by the findings of CheckMate 026[62] and CheckMate 227.[24] After the failure of CheckMate026, the researchers retrospectively stratified the population based on TMB and found that it was a potential biomarker for improved outcomes from nivolumab treatment. TMB analysis might be useful in candidate selection for immunotherapy plus chemotherapy combination. Nivolumab plus ipilimumab combination was also considered as a possible treatment algorithm for NSCLC with high TMB levels (≥10 mut/Mb). However, the application of TMB is controversial. Only 57.7% of the samples were adequate for TMB testing in Checkmate 227 trial.[63] Furthermore, TMB partially reflects tumor neoantigens, which is more closely related to the response rate of the treatment. The Memorial Sloan Kettering Cancer Center team studied the clinical and genomic data of 1662 advanced cancer patients treated with ICIs and 5371 patients not treated with ICIs. They found that even though high TMB is a predictor of efficacy, it is not a prognostic factor.[64] In addition, the long time and high costs associated with TMB and the decision regarding the use of whole-exome sequencing (WES) or panel detection are issues that need to be addressed in future research.

Recently, a novel blood-based assay evaluating TMB in plasma (bTMB) was retrospectively assessed in OAK and POPLAR trials.[65] It was discerned that bTMB reproducibly identified patients with significantly improved PFS in second-line atezolizumab treatment. In the B-F1RST trial of atezolizumab as well as in the MYSTIC[29] trial of durvalumab plus tremelimumab, bTMB was noted to possess clinical value and was found to be more feasible than TMB.

Some studies based on the affinity of new antigens to major histocompatibility complex (MHC) or the similarity of new antigens to known antigens and the machine learning of second-generation sequencing have introduced new antigen prediction models.[66,67] Intra-tumoral heterogeneity (ITH), an uneven distribution of genomic diversification in tumor, was also confirmed as a biomarker for immunotherapy efficacy prediction. However, these potential biomarkers are yet to be tested in clinical practice.

Owing to the complexity of tumor-immune interactions, intra-tumoral heterogeneity, and the cutoff value for “high” TMB definition, TMB alone does not explicitly identify all NSCLC patients who are likely to benefit from immunotherapy. The dynamic monitoring of TMB or neoantigen fitness models directly based on MHC binding affinity might be the future trend in response prediction.

Tumor infiltrating lymphocytes (TILs)

TILs are infiltrating ICs in the TME that mediate anti-tumor immune response.[68] According to ICs infiltration, tumors can be divided into inflamed and non-inflamed types. The presence of TILs, high levels of interferon (IFN) γ-producing CD8+ T-cells, and positive PD-L1 expression in TILs are characteristics of inflamed tumors.[69] Several researchers have reported that TILs are predictive biomarkers in immunotherapy. Tumeh et al[70] established the correlation between the efficacy of PD-1 inhibitor and TILs in melanoma. In serially sampled tumors, patients responding well to PD-1 inhibitor showed upregulated levels of intratumoral CD8+ T-cells rather than CD4+ T-cells, which is directly correlated with a reduction in tumor size. In the OAK trial, PD-L1 expression in TILs was determined to be an independent predictor of benefitting from atezolizumab.[71] Teng et al[72] stratified tumors into four types based on the expression of TILs and PD-L1 as follows: type I: TILs+ PD-L1+; type II: TILs+ PD-L1−; type III: TILs− PD-L1+; and type IV: TILs− PD-L1−. Preliminary studies have reported that type I is more likely to benefit from PD-1/PD-L1 inhibitors. Notably, TILs have not been proposed as a biomarker for choosing the combination regimen of immunotherapy. Currently, a preclinical study has found that TILs reflect radiation-induced DNA damage in hepatocellular carcinoma.[73] In lung cancer mice models, radiotherapy could also enhance the function of TILs, which was strengthened in combination with anti-PD-L1.[74] In the future, more supporting data are still needed.

Genomics feature

Microsatellite instability and mismatch repair (MMR) deficiency are also emerging as indicators for treatment with ICIs. Genetic alterations encoding MMR proteins such as MutS Homolog 2 (MSH2), MutS Homolog 6 (MSH6), and postmeiotic segregation increased 2 (PMS2), which lead to DNA replication errors, have been proven to influence the response to immunotherapy.[75] Similarly, other DNA repair alterations such as DNA polymerase epsilon (POLE) and DNA polymerase delta (POLD) are also associated with the clinical benefit of immunotherapy. So far, as for immunotherapy combination, there is still a lack of direct evidence demonstrating the relationship between genomics feature and treatment efficacy.[76]

On the contrary, serine/threonine kinase 11/serine-threonine kinase B1 (STK11/LKB1) and Janus kinase 1/2 (JAK1/2) mutations suggest primary resistance to ICIs. Skoulidis et al[77] reported that ORR to PD-1 inhibitors differed among the STK11/LKB1 (KL), TP53 (KP), and KRAS-only (K-only) subgroups of KRAS-mutant lung adenocarcinoma. In the Checkmate-057 clinical trial, the KL group had shorter PFS (P < 0.001) and OS (P = 0.002) than the other groups. These results allude that STK11/LKB1 mutations are the main driving factors for primary resistance to PD-1 inhibitors in KRAS-mutant lung adenocarcinoma. Certain studies[78,79] have also found that the somatic JAK1/2 mutations in TCs were responsible for the inability to respond to IFN-γ as well as the absence of reactive PD-L1 expression.

Gut microbiome

The gut microbiome has been proposed to have an impact on the development of cancer and systemic immunity.[80,81] Even though the specific mechanism is not yet verified, increasing evidence shows that the microbiome could be a potential biomarker for the therapeutic outcome. Gopalakrishnan et al[82] deduced that the diversity of gut microbiome was positively correlated with the response to immunotherapy in melanoma patients, and this finding was also verified in a mouse model. Mice raised in a germ-free environment did not respond well to anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4 therapy.[83] Even though no evidence was shown in immunotherapy-based combination in NSCLC, recently, a study of renal cell carcinoma (RCC) observed that either in nivolumab monotherapy (77%) or nivolumab plus ipilimumab (23%), greater microbial diversity was associated with clinical benefit.[84] In general, the abundance of microbiome could be a potential indicator for response to ICI-based therapy.

There are currently no effective markers for combination therapy prediction to choose which patients are more suitable for immunotherapy-based combination. Therefore, the choice of combination regimen is based on predictive indicators of immune monotherapy. The latest perspective is related to the concept of the landmark “Landscape,” which covers many aspects such as PD-L1, TMB, tumor neoantigen, genetic mutation, host germline genome, TME, and gut microbiome.[85] In conclusion, a single biomarker is insufficient to differentiate the responders from the non-responders. Emerging biomarkers are needed to refine the selection process. Furthermore, the development of comprehensive predictive models involving a combination of different components is the need of the hour.

Treatment of special populations

EGFR/ALK alterations

Most prospective clinical studies have excluded patients with EGFR mutations or ALK rearrangements, and only limited trials have included such patients. Among them, IMpower150[33,86] exposed that patients with non-squamous advanced NSCLC, including subgroups with EGFR/ALK mutations, could benefit from ABCP. The study asserted the potential value of anti-angiogenetic agents in this setting. KEYNOTE-789 is an ongoing clinical trial investigating a combination of pembrolizumab plus chemotherapy after the progression of first-line TKIs in NSCLC patients with EGFR mutations. However, the initial results have been disappointing.

Therefore, current evidence mostly suggests that ICIs are not suitable for NSCLC patients with EGFR/ALK alterations. Even though the IMpower150 trial found that the combination of anti-angiogenetic agents and ICIs might exhibit synergistic anti-tumor activity in this population, it could also result in fatal adverse reactions. Therefore, for these patients, TKI monotherapy should be the first-line treatment. ICIs and combinations of ICIs and TKIs are not recommended.[39] If the TKI treatment fails or if the patient is not able to tolerate its adverse effects, it is imperative to detect the PD-L1 expression, TMB levels, and tumor immune microenvironment. ICI monotherapy or combination of ICIs with chemotherapy and anti-angiogenic drugs could be considered for NSCLC patients with high PD-L1 expression.[87]

Elderly patients

The clinical benefit of immunotherapy in elderly patients is still controversial.[88] In the subgroup analysis of IMpower132 trial, the PFS was comparable between the older (≥65 years) and younger (<65 years) NSCLC patients receiving immunotherapy plus chemotherapy (HR: 0.55 vs. 0.63); however, the OS was higher in the older patients than that in the younger patients (HR: 0.71 vs. 0.89).[89] A meta-analysis involving 5265 patients explored the differential activity of immunotherapy in four histological settings. When 65 to 70 years was used as the cut-off value, the older and younger groups were found to receive comparable benefits.[90] A previous study reported that immunosenescence (a measure of immunological age), rather than chronological age is more closely associated with poor response of ICIs.[91]

In summary, the use of combination therapy in elderly patients is still controversial. However, the treatment could offer certain benefits for elderly patients with a good general status, and age should not be the only factor in deciding whether immunotherapy should be administered.

Liver metastasis

Advanced NSCLC with liver metastasis is associated with unfavorable prognosis[92] and poor response in ICI monotherapy as well as in combinations of ICIs and chemotherapy.[93] This result could be attributed to the fact that the liver is the primary site of T cell activation and that incomplete activation could lead to abortive activation, exhaustion, and early death of T cells.[94] The presence of liver metastasis is also correlated with reduced marginal infiltrating CD8+ T cells, which sheds light on the potential causes for these disappointing outcomes.[95] However, the combination of bevacizumab, atezolizumab, and chemotherapy in the IMpower150 trial improved the survival in this population, suggesting the key role of anti-angiogenics in patients with liver metastases. Such patients do not constitute the dominant group for immunotherapy. When compared with chemotherapy, immunotherapy combined with anti-angiogenics is a better choice for this population.

Treatment-related adverse events

With the widespread application of ICIs, irAEs have gradually attracted the attention of the researchers. Numerous large-scale clinical trials have reported that the incidence of irAEs is 60% to 80%, and it is similar in combination strategies.[56,96] In the clinical settings, the irAEs depend on the toxicity profiles of the ICIs and originate from the non-specific activation of the immune system.[97,98] The adverse reactions can occur in any organ or tissue and chiefly involve the skin, gastrointestinal tract, endocrine glands, liver, and lungs.[99,100] Even though irAEs are generally manageable (grade 1–2), some serious adverse reactions such as immune-related pneumonia, immune-interstitial nephritis, and immune-related myocarditis could be life-threatening.

Checkpoint inhibitors-associated pneumonitis (CIP), a kind of lung injury caused by ICIs. CIP has differing clinical, imaging, and pathological features and is the main irAE of grade ≥3. A meta-analysis of ICI treatment combined with chemotherapy showed that the relative risk of CIP for the combined treatment was 2.37 (95% CI: 1.27–4.32, P = 0.007).[101] When compared with ICI monotherapy, the combined treatment with PD-1 and CTLA-4 inhibitors was linked to an increased total incidence of CIP (6.6% vs. 1.6%, P < 0.001) and severe CIP (1.5% vs. 0.2%, P = 0.001).[102] The combined strategy of immunotherapy and targeted therapy can also exacerbate the incidence of CIP. In a phase Ib trial of durvalumab and osimertinib for NSCLC treatment, up to 38% of the patients developed CIP, of which the incidence of severe CIP was 15%.[103]

Although the NSCLC patients benefit from immunotherapy combinations, the upregulated incidence of CIP requires the attention of clinicians. In general, with regard to irAEs reported in the literature, the identification, monitoring, and follow-up of adverse reactions should be carried out throughout the process.

Selection of the optimal combination strategy

Several combination algorithms have been successful in all pathological types of metastatic NSCLC, and most of them have been approved by the FDA. However, in different settings, ICI agents and/or chemotherapy regimens are not exactly the same. The selection criteria for combined therapy are also not completely clear.

First, the existing combination strategies are simply a combination of several agents, leading to just modest ORR in some trials. The optimal kind of drug, dose, and sequence should be chosen to identify the optimized model of the combinations in every setting. Second, it is necessary to consider issues such as adverse reactions. Although irAEs are mostly tolerable, severe irAEs are possible and they can be life-threatening. Even though there is little evidence to suggest that combination strategies are associated with increased mortality or severity of toxicity, clinical benefit and treatment-related toxicity should be balanced. Third, most of NSCLC patients enrolled in the trials were supported by the company, and hence, the expenses of the combination strategies are not taken into account. The current strategies may be suitable only for patients who are economically sound. Therefore, the cost effectiveness factor needs to be considered for routine use of the treatment in the future.

The current clinical trials have focused on the development of novel agents or combination algorithms for immunotherapy. However, the most suitable drugs and dosage, the sequence and the best application time of immunotherapy remain unestablished. In the later stages, more head-to-head phase III trials with large sample sizes are needed to identify the most effective combination strategy. If the toxicity is acceptable, the optimal combination strategy can enhance innate as well as adaptive immunity.

Future challenges

Immunotherapy has truly revolutionized the treatment approach to NSCLC. As per the existing research evidence, immune monotherapy cannot completely satisfy the needs of NSCLC patients. However, immunotherapy combinations are expected to become the trend in the future.

In recent years, treatment algorithms such as immunotherapy combined with chemotherapy, anti-angiogenesis therapy, and targeted therapy have witnessed rapid development. In addition, immunotherapy combined with other treatments such as interventional therapy, ablational therapy, and vaccination is also likely to become the key approaches in tumor management.

Herein, we have proposed four main challenges for the development of immune combined therapy. The first one addresses how to improve the efficiency of immunotherapy. Currently, the optimal combined algorithms, such as the best combined immunotherapy regimen, the dosage, and the best application time, require further optimization. It is well known that the prognosis is significantly better for patients showing efficacy during the early period of the application of the immune combination therapy, such that it has become necessary to promote early efficacy manifestation. Meanwhile, the development and research of new immunomodulatory agents, including CAR-T-cell/CAR-natural killer-cell therapies, should be accelerated to break through the bottleneck of immune combination therapy. In this aspect, more numbers of pre-clinical and clinical trials are needed in the future to promote the efficacy of immunotherapy for lung cancer.

The second one addresses how to improve the preciseness of immunotherapy. Although combined algorithms appear to be the future trend, the existence of ITH poses a considerable challenge in the implementation of precision oncology. The PD-L1 expression levels and TMB have been routinely assessed in clinical trials, which should actually be the foundation of the biomarker exploration. It is, therefore, necessary to introduce the concept of “Landscape,” which covers multiple aspects such as PD-L1, TMB, tumor neoantigen, genetic feature, TME, and imaging in order to explore the individualized markers of immune efficacy.

The third one addresses measures to prolong the benefit of immunotherapy. The endogenous and exogenous mechanisms through which the immune combination can regulate multiple aspects to overcome drug resistance remain unclear. Ascertaining the primary drug-resistant population is also difficult. On the one hand, a pre-clinical study should explore the in-depth understanding of the mechanisms of drug resistance during immune combination therapy. On the other hand, clinical practice needs to delay or overcome drug resistance by bringing immunotherapy forward to the first-line in a combined fashion, by turning cold tumors into hot ones.

Finally, we addressed the mechanism to make immunotherapy more secure. When compared with the use of chemotherapy and targeted therapies, the specific damage caused by immunotherapy remains unclear. It may affect the multiple systems throughout the body (eg, the heart, thyroid, small intestine, and pituitary) and some irAEs of grade ≥3 can progress rapidly within a short period of time. Therefore, irAEs require close attention and effective clinical management methods are urgently needed for irAEs. In the future, avoiding the occurrence of serious irAEs, adopting an individualized approach to manage irAEs in a better way, and timing the “re-challenge” after the control of irAEs will be the hotspots of clinical concern.

The future of NSCLC treatment is thus immune combination. However, it is undeniable that the exploration of immunotherapy for NSCLC is still in its infancy. This industry needs innovation, integration, and transformation to promote the continuous advancement of immune combination. We are confident that once the mystery shrouding immunotherapy is unveiled, the adoption of this strategy will mark the beginning of a new era.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81802301 and 81772500) and the Social Development Foundation of China (No. BE2019719).

Conflicts of interest

None.

Footnotes

How to cite this article: Li CL, Song Y. Combination strategies of immunotherapy in non-small cell lung cancer: facts and challenges. Chin Med J 2021;134:1908–1919. doi: 10.1097/CM9.0000000000001610

References

  • 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
  • 2.Lazzari C, Karachaliou N, Gregorc V, Bulotta A, Gonzalez-Cao M, Verlicchi A, et al. Second-line therapy of squamous non-small cell lung cancer: an evolving landscape. Expert Rev Respir Med 2017; 11:469–479. doi: 10.1080/17476348.2017.1326822. [DOI] [PubMed] [Google Scholar]
  • 3.Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol 2014; 27:16–25. doi: 10.1016/j.coi.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pardoll DM. Immunology beats cancer: a blueprint for successful translation. Nat Immunol 2012; 13:1129–1132. doi: 10.1038/ni.2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Shi T, Ma Y, Yu L, Jiang J, Shen S, Hou Y, et al. Cancer immunotherapy: a focus on the regulation of immune checkpoints. Int J Mol Sci 2018; 19:1389.doi: 10.3390/ijms19051389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Weiss T, Weller M, Roth P. Immunological effects of chemotherapy and radiotherapy against brain tumors. Expert Rev Anticancer Ther 2016; 16:1087–1094. doi: 10.1080/14737140.2016.1229600. [DOI] [PubMed] [Google Scholar]
  • 7.Hato SV, Khong A, de Vries IJ, Lesterhuis WJ. Molecular pathways: the immunogenic effects of platinum-based chemotherapeutics. Clin Cancer Res 2014; 20:2831–2837. doi: 10.1158/1078-0432.CCR-13-3141. [DOI] [PubMed] [Google Scholar]
  • 8.Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M, et al. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev 2011; 30:61–69. doi: 10.1007/s10555-011-9273-4. [DOI] [PubMed] [Google Scholar]
  • 9.Qiao M, Jiang T, Ren S, Zhou C. Combination strategies on the basis of immune checkpoint inhibitors in non-small-cell lung cancer: where do we stand? Clin Lung Cancer 2018; 19:1–11. doi: 10.1016/j.cllc.2017.06.005. [DOI] [PubMed] [Google Scholar]
  • 10.Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T, et al. Chemotherapy induces programmed cell death-ligand 1 Overexpression via the nuclear factor-κB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res 2015; 75:5034–5045. doi: 10.1158/0008-5472.CAN-14-3098. [DOI] [PubMed] [Google Scholar]
  • 11.McDonnell AM, Lesterhuis WJ, Khong A, Nowak AK, Lake RA, Currie AJ, et al. Tumor-infiltrating dendritic cells exhibit defective cross-presentation of tumor antigens, but is reversed by chemotherapy. Eur J Immunol 2015; 45:49–59. doi: 10.1002/eji.201444722. [DOI] [PubMed] [Google Scholar]
  • 12.Liu WM, Fowler DW, Smith P, Dalgleish AG. Pre-treatment with chemotherapy can enhance the antigenicity and immunogenicity of tumours by promoting adaptive immune responses. Br J Cancer 2010; 102:115–123. doi: 10.1038/sj.bjc.6605465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Langer CJ, Gadgeel SM, Borghaei H, Papadimitrakopoulou VA, Patnaik A, Powell SF, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol 2016; 17:1497–1508. doi: 10.1016/S1470-2045(16)30498-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Awad MM, Gadgeel SM, Borghaei H, Patnaik A, Yang JC, Powell SF, et al. Long-term overall survival from KEYNOTE-021 cohort G: pemetrexed and carboplatin with or without pembrolizumab as first-line therapy for advanced nonsquamous NSCLC. J Thorac Oncol 2021; 16:162–168. doi: 10.1016/j.jtho.2020.09.015. [DOI] [PubMed] [Google Scholar]
  • 15.Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018; 378:2078–2092. doi: 10.1056/NEJMoa1801005. [DOI] [PubMed] [Google Scholar]
  • 16.Gadgeel S, Rodríguez-Abreu D, Speranza G, Esteban E, Felip E, Dómine M, et al. Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol 2020; 38:1505–1517. doi: 10.1200/JCO.19.03136. [DOI] [PubMed] [Google Scholar]
  • 17.Paz-Ares L, Vicente D, Tafreshi A, Robinson A, Soto Parra H, Mazières J, et al. A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous NSCLC: protocol-specified final analysis of KEYNOTE-407. J Thorac Oncol 2020; 15:1657–1669. doi: 10.1016/j.jtho.2020.06.015. [DOI] [PubMed] [Google Scholar]
  • 18.West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, Conter HJ, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20:924–937. doi: 10.1016/S1470-2045(19)30167-6. [DOI] [PubMed] [Google Scholar]
  • 19.Jotte R, Cappuzzo F, Vynnychenko I, Stroyakovskiy D, Rodríguez-Abreu D, Hussein M, 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 2020; 15:1351–1360. doi: 10.1016/j.jtho.2020.03.028. [DOI] [PubMed] [Google Scholar]
  • 20.Nishio M, Barlesi F, West H, Ball S, Bordoni R, Cobo M, et al. Atezolizumab plus chemotherapy for first-line treatment of non-squamous non-small cell lung cancer: results from the randomized phase III IMpower132 trial. J Thorac Oncol 2020; 16:653–664. doi: 10.1016/j.jtho.2020.11.025. [DOI] [PubMed] [Google Scholar]
  • 21.Arrieta O, Barrón F, Ramírez-Tirado LA, Zatarain-Barrón ZL, Cardona AF, Díaz-García D, et al. Efficacy and safety of pembrolizumab plus docetaxel vs docetaxel alone in patients with previously treated advanced non-small cell lung cancer: the PROLUNG phase 2 randomized clinical trial. JAMA Oncol 2020; 6:856–864. doi: 10.1001/jamaoncol.2020.0409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hellmann MD, Rizvi NA, Goldman JW, Gettinger SN, Borghaei H, Brahmer JR, 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 2017; 18:31–41. doi: 10.1016/S1470-2045(16)30624-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F, Ahuja A, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 2018; 33:843–852.e4. doi: 10.1016/j.ccell.2018.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med 2019; 381:2020–2031. doi: 10.1056/NEJMoa1910231. [DOI] [PubMed] [Google Scholar]
  • 25.Reck M, Ciuleanu TE, Lee JS, Schenker M, Audigier-Valette C, Zurawski B, 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 2021; 16:665–676. doi: 10.1016/j.jtho.2020.12.019. [DOI] [PubMed] [Google Scholar]
  • 26.Ramalingam SS, Ciuleanu TE, Pluzanski A, Lee J-S, Schenker M, Bernabe Caro R, et al. Nivolumab + ipilimumab versus platinum-doublet chemotherapy as first-line treatment for advanced non-small cell lung cancer: three-year update from CheckMate 227 Part 1. J Clin Oncol 2020; 38: 15_suppl: 9500.doi: 10.1200/JCO.2020.38.15_suppl.9500. [Google Scholar]
  • 27.Paz-Ares L, Ciuleanu TE, Cobo M, Schenker M, Zurawski B, Menezes J, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, phase 3 trial. Lancet Oncol 2021; 22:198–211. doi: 10.1016/S1470-2045(20)30641-0. [DOI] [PubMed] [Google Scholar]
  • 28.Boyer M, Şendur M, Rodríguez-Abreu D, Park K, Lee DH, Çiçin I, et al. Pembrolizumab plus ipilimumab or placebo for metastatic non-small-cell lung cancer with PD-L1 tumor proportion score ≥ 50%: randomized, double-blind phase III KEYNOTE-598 study. J Clin Oncol 2021; doi: 10.1200/JCO.20.03579. [DOI] [PubMed] [Google Scholar]
  • 29.Rizvi NA, Cho BC, Reinmuth N, Lee KH, Luft A, Ahn MJ, 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 2020; 6:661–674. doi: 10.1001/jamaoncol.2020.0237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. AstraZeneca. Update on the Phase III NEPTUNE Trial of Imfinzi Plus Tremelimumab in Stage IV Non-small-cell Lung Cancer. 2019 Available from: https://www.astrazeneca.com/media-centre/press-releases/2019/update-on-the-phase-iii-neptune-trial-of-imfinzi-plus-tremelimumab-in-stage-iv-non-small-cell-lung-cancer-21082019.html. [Google Scholar]
  • 31.Kowalski DM, Reinmuth N, Orlov SV, Fischer JR, Sugawara S, Mandziuk S, et al. ARCTIC: durvalumab + tremelimumab and durvalumab monotherapy vs SoC in ≥ 3L advanced NSCLC treatment. Ann Oncol 2018; 29:viii493–viii494. doi: 10.1093/annonc/mdy292.001. [Google Scholar]
  • 32.Hegde PS, Wallin JJ, Mancao C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol 2018; 52:117–124. doi: 10.1016/j.semcancer.2017.12.002. [DOI] [PubMed] [Google Scholar]
  • 33.Reck M, Mok T, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, 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 2019; 7:387–401. doi: 10.1016/S2213-2600(19)30084-0. [DOI] [PubMed] [Google Scholar]
  • 34.Hui R, Nishio M, Reck M, Rodriguez-Abreu D, Fouad TM, Flaim D, et al. Randomized, double-blind, phase 3 trial of first-line pembrolizumab + platinum doublet chemotherapy (chemo) ± lenvatinib in patients (pts) with metastatic nonsquamous non–small-cell lung cancer (NSCLC): LEAP-006. J Clin Oncol 2019; 37: 15_suppl: TS9118.doi: 10.1200/JCO.2019.37.15_suppl.TPS9118. [Google Scholar]
  • 35.Shiraishi Y, Daga H, Ikeda S, Hata A, Mizutani H, Sakamoto T, et al. A multicenter, open label, randomized phase III study of atezolizumab with platinum-pemetrexed and with or without bevacizumab for patients with advanced nonsquamous non-small cell lung cancer (WJOG11218L APPLE study). J Clin Oncol 2019; 37: 15_suppl: TS9125.doi: 10.1200/JCO.2019.37.15_suppl.TPS9125. [Google Scholar]
  • 36.Yang JC, Gadgeel SM, Sequist LV, Wu CL, Papadimitrakopoulou VA, Su WC, et al. Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol 2019; 14:553–559. doi: 10.1016/j.jtho.2018.11.028. [DOI] [PubMed] [Google Scholar]
  • 37.Gettinger S, Hellmann MD, Chow L, Borghaei H, Antonia S, Brahmer JR, et al. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J Thorac Oncol 2018; 13:1363–1372. doi: 10.1016/j.jtho.2018.05.015. [DOI] [PubMed] [Google Scholar]
  • 38.Gibbons DL, Chow LQ, Kim DW, Kim SW, Yeh T, Song X, et al. 57O Efficacy, safety and tolerability of MEDI4736 (durvalumab [D]), a human IgG1 anti-programmed cell death-ligand-1 (PD-L1) antibody, combined with gefitinib (G): a phase I expansion in TKI-naïve patients (pts) with EGFR mutant NSCLC. J Thorac Oncol 2016; 11:S79.doi: 10.1016/s1556-0864(16)30171-x. [Google Scholar]
  • 39.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 2017; 16:465–469. doi: 10.1080/14740338.2017.1300656. [DOI] [PubMed] [Google Scholar]
  • 40.Spigel DR, Reynolds C, Waterhouse D, Garon EB, Chandler J, Babu S, 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 2018; 13:682–688. doi: 10.1016/j.jtho.2018.02.022. [DOI] [PubMed] [Google Scholar]
  • 41.Kim D-W, Gadgeel SM, Gettinger SN, Riely GJ, Oxnard GR, Mekhail T, et al. Safety and clinical activity results from a phase Ib study of alectinib plus atezolizumab in ALK+ advanced NSCLC (aNSCLC). J Clin Oncol 2018; 36: 15_suppl: 9009.doi: 10.1200/JCO.2018.36.15_suppl.9009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer 2020; 20:203–217. doi: 10.1038/s41568-020-0246-1. [DOI] [PubMed] [Google Scholar]
  • 43.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 2017; 377:1919–1929. doi: 10.1056/NEJMoa1709937. [DOI] [PubMed] [Google Scholar]
  • 44.Gray JE, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC-update from PACIFIC. J Thorac Oncol 2020; 15:288–293. doi: 10.1016/j.jtho.2019.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Durm GA, Jabbour SK, Althouse SK, Liu Z, Sadiq AA, Zon RT, et al. A phase 2 trial of consolidation pembrolizumab following concurrent chemoradiation for patients with unresectable stage III non-small cell lung cancer: Hoosier Cancer Research Network LUN 14-179. Cancer 2020; 126:4353–4361. doi: 10.1002/cncr.33083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jabbour SK, Lee KH, Frost N, Kowalski D, Breder VV, Pollock T, et al. Phase II study of pembrolizumab (pembro) plus platinum doublet chemotherapy and radiotherapy as first-line therapy for unresectable, locally advanced stage III NSCLC: KEYNOTE-799. J Clin Oncol 2020; 38: 15_suppl: 9008.doi: 10.1200/JCO.2020.38.15_suppl.9008. [Google Scholar]
  • 47.Rocco D, Gregorc V, Della Gravara L, Lazzari C, Palazzolo G, Gridelli C. New immunotherapeutic drugs in advanced non-small cell lung cancer (NSCLC): from preclinical to phase I clinical trials. Expert Opin Investig Drugs 2020; 29:1005–1023. doi: 10.1080/13543784.2020.1793956. [DOI] [PubMed] [Google Scholar]
  • 48.Donini C, D’Ambrosio L, Grignani G, Aglietta M, Sangiolo D. Next generation immune-checkpoints for cancer therapy. J Thorac Dis 2018; 10:S1581–S1601. doi: 10.21037/jtd.2018.02.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Houssaini MS, Damou M, Ismaili N. Advances in the management of non-small cell lung cancer (NSCLC): a new practice changing data from ASCO 2020 annual meeting. Cancer Treat Res Commun 2020; 25:100239.doi: 10.1016/j.ctarc.2020.100239. [DOI] [PubMed] [Google Scholar]
  • 50.Li A, Barsoumian HB, Schoenhals JE, Cushman TR, Caetano MS, Wang X, et al. Indoleamine 2,3-dioxygenase 1 inhibition targets anti-PD1-resistant lung tumors by blocking myeloid-derived suppressor cells. Cancer Lett 2018; 431:54–63. doi: 10.1016/j.canlet.2018.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Le Naour J, Galluzzi L, Zitvogel L, Kroemer G, Vacchelli E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2020; 9:1777625.doi: 10.1080/2162402x.2020.1777625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Qu J, Mei Q, Chen L, Zhou J. Chimeric antigen receptor (CAR)-T-cell therapy in non-small-cell lung cancer (NSCLC): current status and future perspectives. Cancer Immunol Immunother 2021; 70:619–631. doi: 10.1007/s00262-020-02735-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Saltos AN, Tanvetyanon T, Creelan BC, Shafique MR, Antonia SJ, Haura EB, et al. Phase II randomized trial of first-line pembrolizumab and vorinostat in patients with metastatic NSCLC (mNSCLC). J Clin Oncol 2020; 38: 15_suppl: 9567.doi: 10.1200/JCO.2020.38.15_suppl.9567. [Google Scholar]
  • 54.Ramalingam SS, Thara E, Rao S, Awad MM, Dowlati A, Haque B, et al. 1268P JASPER: efficacy and safety of first-line (1L) niraparib plus a programmed death receptor 1 inhibitor (PD-1i) in patients with advanced non-small cell lung cancer (NSCLC). Ann Oncol 2020; 31:S819–S820. doi: 10.1016/j.annonc.2020.08.1582. [Google Scholar]
  • 55.Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, 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. doi: 10.1016/S0140-6736(15)01281-7. [DOI] [PubMed] [Google Scholar]
  • 56.Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015; 373:123–135. doi: 10.1056/NEJMoa1504627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jotte RM, Cappuzzo F, Vynnychenko I, Stroyakovskiy D, Abreu DR, Hussein MA, et al. IMpower131: primary PFS and safety analysis of a randomized phase III study of atezolizumab + carboplatin + paclitaxel or nab-paclitaxel vs carboplatin + nab-paclitaxel as 1L therapy in advanced squamous NSCLC. J Clin Oncol 2018; 36: 18_suppl: LBA9000.doi: 10.1200/JCO.2018.36.18_suppl.LBA9000. [Google Scholar]
  • 58.Boyer M, Şendur MAN, Rodríguez-Abreu D, Park K, Lee DH, Çiçin I, et al. Pembrolizumab plus ipilimumab or placebo for metastatic non-small-cell lung cancer with PD-L1 tumor proportion score ≥ 50%: randomized, double-blind phase III KEYNOTE-598 Study. J Clin Oncol 2021; JCO2003579.doi: 10.1200/JCO.20.03579. [DOI] [PubMed] [Google Scholar]
  • 59.Tsao MS, Kerr KM, Kockx M, Beasley MB, Borczuk AC, Botling J, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: results of blueprint phase 2 project. J Thorac Oncol 2018; 13:1302–1311. doi: 10.1016/j.jtho.2018.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, 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 2017; 12:208–222. doi: 10.1016/j.jtho.2016.11.2228. [DOI] [PubMed] [Google Scholar]
  • 61.Ilie M, Long-Mira E, Bence C, Butori C, Lassalle S, Bouhlel L, et al. Comparative study of the PD-L1 status between surgically resected specimens and matched biopsies of NSCLC patients reveal major discordances: a potential issue for anti-PD-L1 therapeutic strategies. Ann Oncol 2016; 27:147–153. doi: 10.1093/annonc/mdv489. [DOI] [PubMed] [Google Scholar]
  • 62.Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med 2017; 376:2415–2426. doi: 10.1056/NEJMoa1613493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.High TMB predicts immunotherapy benefit. Cancer Discov 2018; 8:668.doi: 10.1158/2159-8290.CD-NB2018-048. [DOI] [PubMed] [Google Scholar]
  • 64.Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019; 51:202–206. doi: 10.1038/s41588-018-0312-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gandara DR, Paul SM, Kowanetz M, Schleifman E, Zou W, Li Y, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 2018; 24:1441–1448. doi: 10.1038/s41591-018-0134-3. [DOI] [PubMed] [Google Scholar]
  • 66.Łuksza M, Riaz N, Makarov V, Balachandran VP, Hellmann MD, Solovyov A, et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017; 551:517–520. doi: 10.1038/nature24473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kim S, Kim HS, Kim E, Lee MG, Shin EC, Paik S, et al. Neopepsee: accurate genome-level prediction of neoantigens by harnessing sequence and amino acid immunogenicity information. Ann Oncol 2018; 29:1030–1036. doi: 10.1093/annonc/mdy022. [DOI] [PubMed] [Google Scholar]
  • 68.Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016; 22:1865–1874. doi: 10.1158/1078-0432.CCR-15-1507. [DOI] [PubMed] [Google Scholar]
  • 69.Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348:56–61. doi: 10.1126/science.aaa8172. [DOI] [PubMed] [Google Scholar]
  • 70.Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014; 515:568–571. doi: 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, 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. doi: 10.1016/S0140-6736(16)32517-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015; 75:2139–2145. doi: 10.1158/0008-5472.CAN-15-0255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Huang Y, Sheng H, Xiao Y, Hu W, Zhang Z, Chen Y, et al. Wnt/β-catenin inhibitor ICG-001 enhances the antitumor efficacy of radiotherapy by increasing radiation-induced DNA damage and improving tumor immune microenvironment in hepatocellular carcinoma. Radiother Oncol 2021; S0167-8140(21)06621-4.doi: 10.1016/j.radonc.2021.06.034. [DOI] [PubMed] [Google Scholar]
  • 74.Chen JL, Pan CK, Huang YS, Tsai CY, Wang CW, Lin YL, et al. Evaluation of antitumor immunity by a combination treatment of high-dose irradiation, anti-PDL1, and anti-angiogenic therapy in murine lung tumors. Cancer Immunol Immunother 2021; 70:391–404. doi: 10.1007/s00262-020-02690-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response to immune checkpoint blockade. Nat Med 2019; 25:389–402. doi: 10.1038/s41591-019-0382-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Campbell BB, Light N, Fabrizio D, Zatzman M, Fuligni F, de Borja R, et al. Comprehensive analysis of hypermutation in human cancer. Cell 2017; 171:1042–1056.e10. doi: 10.1016/j.cell.2017.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov 2018; 8:822–835. doi: 10.1158/2159-8290.CD-18-0099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov 2017; 7:188–201. doi: 10.1158/2159-8290.CD-16-1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016; 375:819–829. doi: 10.1056/NEJMoa1604958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer 2017; 17:271–285. doi: 10.1038/nrc.2017.13. [DOI] [PubMed] [Google Scholar]
  • 81.Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol 2017; 15:465–478. doi: 10.1038/nrmicro.2017.44. [DOI] [PubMed] [Google Scholar]
  • 82.Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359:97–103. doi: 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350:1079–1084. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Salgia NJ, Bergerot PG, Maia MC, Dizman N, Hsu J, Gillece JD, et al. Stool microbiome profiling of patients with metastatic renal cell carcinoma receiving anti-PD-1 immune checkpoint inhibitors. Eur Urol 2020; 78:498–502. doi: 10.1016/j.eururo.2020.07.011. [DOI] [PubMed] [Google Scholar]
  • 85.Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019; 19:133–150. doi: 10.1038/s41568-019-0116-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med 2018; 378:2288–2301. doi: 10.1056/NEJMoa1716948. [DOI] [PubMed] [Google Scholar]
  • 87.Garassino MC, Cho BC, Kim JH, Mazières J, Vansteenkiste J, Lena H, 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 2018; 19:521–536. doi: 10.1016/S1470-2045(18)30144-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Remon J, Vilariño N, Reguart N. Immune checkpoint inhibitors in non-small cell lung cancer (NSCLC): approaches on special subgroups and unresolved burning questions. Cancer Treat Rev 2018; 64:21–29. doi: 10.1016/j.ctrv.2018.02.002. [DOI] [PubMed] [Google Scholar]
  • 89.Barlesi F, Nishio M, Cobo M, Steele N, Paramonov V, Parente B, et al. IMpower132: Efficacy of atezolizumab (atezo) + carboplatin (carbo)/cisplatin (cis) + pemetrexed (pem) as 1L treatment in key subgroups with stage IV non-squamous non-small cell lung cancer (NSCLC). Annals Oncol 2018; 29: suppl_8: viii743–viii744. doi: 10.1093/annonc/mdy424.066. [Google Scholar]
  • 90.Nishijima TF, Muss HB, Shachar SS, Moschos SJ. Comparison of efficacy of immune checkpoint inhibitors (ICIs) between younger and older patients: a systematic review and meta-analysis. Cancer Treat Rev 2016; 45:30–37. doi: 10.1016/j.ctrv.2016.02.006. [DOI] [PubMed] [Google Scholar]
  • 91.Ferrara R, Mezquita L, Auclin E, Chaput N, Besse B. Immunosenescence and immunecheckpoint inhibitors in non-small cell lung cancer patients: does age really matter? Cancer Treat Rev 2017; 60:60–68. doi: 10.1016/j.ctrv.2017.08.003. [DOI] [PubMed] [Google Scholar]
  • 92.Li J, Zhu H, Sun L, Xu W, Wang X. Prognostic value of site-specific metastases in lung cancer: a population based study. J Cancer 2019; 10:3079–3086. doi: 10.7150/jca.30463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tournoy KG, Thomeer M, Germonpré P, Derijcke S, De Pauw R, Galdermans D, et al. Does nivolumab for progressed metastatic lung cancer fulfill its promises? An efficacy and safety analysis in 20 general hospitals. Lung Cancer 2018; 115:49–55. doi: 10.1016/j.lungcan.2017.11.008. [DOI] [PubMed] [Google Scholar]
  • 94.Horst AK, Neumann K, Diehl L, Tiegs G. Modulation of liver tolerance by conventional and nonconventional antigen-presenting cells and regulatory immune cells. Cell Mol Immunol 2016; 13:277–292. doi: 10.1038/cmi.2015.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tumeh PC, Hellmann MD, Hamid O, Tsai KK, Loo KL, Gubens MA, et al. Liver metastasis and treatment outcome with anti-PD-1 monoclonal antibody in patients with melanoma and NSCLC. Cancer Immunol Res 2017; 5:417–424. doi: 10.1158/2326-6066.CIR-16-0325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, 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. doi: 10.1016/S0140-6736(16)00587-0. [DOI] [PubMed] [Google Scholar]
  • 97.Arnaud-Coffin P, Maillet D, Gan HK, Stelmes JJ, You B, Dalle S, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer 2019; 145:639–648. doi: 10.1002/ijc.32132. [DOI] [PubMed] [Google Scholar]
  • 98.Hoffner B, Leighl NB, Davies M. Toxicity management with combination chemotherapy and programmed death 1/programmed death ligand 1 inhibitor therapy in advanced lung cancer. Cancer Treat Rev 2020; 85:101979.doi: 10.1016/j.ctrv.2020.101979. [DOI] [PubMed] [Google Scholar]
  • 99.Costa R, Carneiro BA, Agulnik M, Rademaker AW, Pai SG, Villaflor VM, et al. Toxicity profile of approved anti-PD-1 monoclonal antibodies in solid tumors: a systematic review and meta-analysis of randomized clinical trials. Oncotarget 2017; 8:8910–8920. doi: 10.18632/oncotarget.13315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.De Velasco G, Je Y, Bossé D, Awad MM, Ott PA, Moreira RB, et al. Comprehensive meta-analysis of key immune-related adverse events from CTLA-4 and PD-1/PD-L1 inhibitors in cancer patients. Cancer Immunol Res 2017; 5:312–318. doi: 10.1158/2326-6066.CIR-16-0237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Huang Y, Fan H, Li N, Du J. Risk of immune-related pneumonitis for PD1/PD-L1 inhibitors: systematic review and network meta-analysis. Cancer Med 2019; 8:2664–2674. doi: 10.1002/cam4.2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol 2017; 35:709–717. doi: 10.1200/JCO.2016.68.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Oxnard GR, Yang JC, Yu H, Kim SW, Saka H, Horn L, et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann Oncol 2020; 31:507–516. doi: 10.1016/j.annonc.2020.01.013. [DOI] [PubMed] [Google Scholar]

Articles from Chinese Medical Journal are provided here courtesy of Wolters Kluwer Health

RESOURCES