Combined Checkpoint Inhibition and Chemotherapy: New Era of 1St-Line Treatment for Non-Small-Cell Lung Cancer
Chongkai Wang,Prakash Kulkarni,Ravi Salgia
DOI: https://doi.org/10.1016/j.omto.2019.02.001
2019-01-01
Abstract:Platinum-based chemotherapy has long been the first-line treatment of choice for metastatic non-small-cell lung cancer (NSCLC) patients who lack targetable gene mutations. The arrival of checkpoint blockade has led to a vast shift in the treatment landscape of NSCLC. Among NSCLC patients with PD-L1 expression in ≥50% of tumor cells, treatment with pembrolizumab leads to a superior progression-free and overall survival compared to platinum-doublet chemotherapy in the first-line setting. Furthermore, the addition of pembrolizumab to standard chemotherapy of pemetrexed and a platinum-based drug resulted in significant longer progression-free survival and overall survival irrespective to PD-L1 expression. In this review, we focus on the molecular rationale for the combination therapy and the results of completed clinical studies. Platinum-based chemotherapy has long been the first-line treatment of choice for metastatic non-small-cell lung cancer (NSCLC) patients who lack targetable gene mutations. The arrival of checkpoint blockade has led to a vast shift in the treatment landscape of NSCLC. Among NSCLC patients with PD-L1 expression in ≥50% of tumor cells, treatment with pembrolizumab leads to a superior progression-free and overall survival compared to platinum-doublet chemotherapy in the first-line setting. Furthermore, the addition of pembrolizumab to standard chemotherapy of pemetrexed and a platinum-based drug resulted in significant longer progression-free survival and overall survival irrespective to PD-L1 expression. In this review, we focus on the molecular rationale for the combination therapy and the results of completed clinical studies. Lung cancer remains one of the leading causes of deaths from cancer worldwide, with ∼80% of all cases presenting as non-small-cell lung cancer (NSCLC).1Torre L.A. Siegel R.L. Ward E.M. Jemal A. Global cancer incidence and mortality rates and trends--an update.Cancer Epidemiol. Biomarkers Prev. 2016; 25: 16-27Crossref PubMed Scopus (2343) Google Scholar, 2Devesa S.S. Bray F. Vizcaino A.P. Parkin D.M. International lung cancer trends by histologic type: male:female differences diminishing and adenocarcinoma rates rising.Int. J. Cancer. 2005; 117: 294-299Crossref PubMed Scopus (623) Google Scholar Currently, the standard first-line treatment for patients with advanced NSCLC without targetable driver mutations is platinum-based, two drug chemotherapy regimens, which achieved a median overall survival of 10.3 months. The addition of bevacizumab to chemotherapy showed modest benefit, with an improved overall survival of 12.3 months.3Sandler A. Gray R. Perry M.C. Brahmer J. Schiller J.H. Dowlati A. Lilenbaum R. Johnson D.H. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer.N. Engl. J. Med. 2006; 355: 2542-2550Crossref PubMed Scopus (5232) Google Scholar Despite extensive investigations, the addition of a third cytotoxic agent to platinum-doublet regimen failed to demonstrate improvement in progression-free survival (PFS) or overall survival (OS) over platinum-doublet chemotherapy alone in various clinical trials.4Azim Jr., H.A. Elattar I. Loberiza Jr., F.R. Azim H. Mok T. Ganti A.K. Third generation triplet cytotoxic chemotherapy in advanced non-small cell lung cancer: a systematic overview.Lung Cancer. 2009; 64: 194-198Abstract Full Text Full Text PDF PubMed Scopus (35) Google Scholar A phase I study of pembrolizumab in patients with advanced NSCLC has shown robust efficacy, including a median OS of 22.3 months in treatment-naive patients and a median OS of 34.9 months in patients whose PD-L1 tumor proportion score (TPS) is ≥50%.5Garon E.B. Rizvi N.A. Hui R. Leighl N. Balmanoukian A.S. Eder J.P. Patnaik A. Aggarwal C. Gubens M. Horn L. et al.KEYNOTE-001 InvestigatorsPembrolizumab for the treatment of non-small-cell lung cancer.N. Engl. J. Med. 2015; 372: 2018-2028Crossref PubMed Scopus (4358) Google Scholar, 6Leighl N.B. Hellmann M.D. Hui R. Costa E.C. Felip E. Ahn M.-J. Eder J.P. Balmanoukian A.S. Aggarwal C. Horn L. et al.KEYNOTE-001: 3-year overall survival for patients with advanced NSCLC treated with pembrolizumab.J. Clin. Oncol. 2017; 35: 9011Crossref Google Scholar This superior efficacy was confirmed by a phase 2 and 3 study, where treatment with pembrolizumab prolonged OS by 2–4 months in PD-L1-positive (TPS ≥ 1%) NSCLC patients who progressed after platinum-based chemotherapy versus standard-of-care treatment.7Herbst R.S. Baas P. Kim D.W. Felip E. Pérez-Gracia J.L. Han J.Y. Molina J. Kim J.H. Arvis C.D. Ahn M.J. et al.Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial.Lancet. 2016; 387: 1540-1550Abstract Full Text Full Text PDF PubMed Scopus (4549) Google Scholar There is now increasing evidence that suggests that the anti-tumor effects of chemotherapy is mediated not only through cytotoxic effects but also through modulating tumor immunity, including inducing immunogenic cell death, enhancing tumor antigen presentation, as well as inhibiting regulatory T cells, and the elimination of myeloid-deprived suppressor cells (MDSCs).8Kepp O. Galluzzi L. Martins I. Schlemmer F. Adjemian S. Michaud M. Sukkurwala A.Q. Menger L. Zitvogel L. Kroemer G. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy.Cancer Metastasis Rev. 2011; 30: 61-69Crossref PubMed Scopus (215) Google Scholar, 9Galluzzi L. Buqué A. Kepp O. Zitvogel L. Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents.Cancer Cell. 2015; 28: 690-714Abstract Full Text Full Text PDF PubMed Scopus (958) Google Scholar, 10Zitvogel L. Galluzzi L. Smyth M.J. Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance.Immunity. 2013; 39: 74-88Abstract Full Text Full Text PDF PubMed Scopus (644) Google Scholar, 11Wang Z. Till B. Gao Q. Chemotherapeutic agent-mediated elimination of myeloid-derived suppressor cells.OncoImmunology. 2017; 6: e1331807Crossref PubMed Scopus (98) Google Scholar Preclinical studies have demonstrated that chemotherapies can stimulate the immune system, causing anti-tumor immunity that contributes to the efficacy of these drugs.12Ramakrishnan R. Assudani D. Nagaraj S. Hunter T. Cho H.I. Antonia S. Altiok S. Celis E. Gabrilovich D.I. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice.J. Clin. Invest. 2010; 120: 1111-1124Crossref PubMed Scopus (360) Google Scholar, 13Yang S. Haluska F.G. Treatment of melanoma with 5-fluorouracil or dacarbazine in vitro sensitizes cells to antigen-specific CTL lysis through perforin/granzyme- and Fas-mediated pathways.J. Immunol. 2004; 172: 4599-4608Crossref PubMed Scopus (76) Google Scholar In addition, early clinical trials for combining PD-1 or PD-L1 blockade with chemotherapy showed superior anti-tumor activity in first-line setting for advanced NSCLCs.14Rizvi N.A. Hellmann M.D. Brahmer J.R. Juergens R.A. Borghaei H. Gettinger S. Chow L.Q. Gerber D.E. Laurie S.A. Goldman J.W. et al.Nivolumab in combination with platinum-based doublet chemotherapy for first-line treatment of advanced non-small-cell lung cancer.J. Clin. Oncol. 2016; 34: 2969-2979Crossref PubMed Scopus (357) Google Scholar, 15Gadgeel S.M. Stevenson J. Langer C.J. Gandhi L. Borghaei H. Patnaik A. Villaruz L.C. Gubens M.A. Hauke R.J. Yang J.C.-H. et al.Pembrolizumab (pembro) plus chemotherapy as front-line therapy for advanced NSCLC: KEYNOTE-021 cohorts A-C.J. Clin. Oncol. 2016; 34: 9016Crossref Google Scholar, 16Liu S.V. Camidge D.R. Gettinger S.N. Giaccone G. Heist R.S. Hodi F.S. Ready N.E. Zhang W. Wallin J. Funke R. et al.Long-term survival follow-up of atezolizumab in combination with platinum-based doublet chemotherapy in patients with advanced non-small-cell lung cancer.Eur. J. Cancer. 2018; 101: 114-122Abstract Full Text Full Text PDF PubMed Scopus (38) Google Scholar These results lend credence to the argument that combination of chemotherapy and immunotherapies is the next step for cancer treatment. Platinum-based chemotherapy has long been the first-line therapy for most patients with metastatic NSCLC without a targetable driver mutation, with a median OS of approximately 12 months.17Leighl N.B. Treatment paradigms for patients with metastatic non-small-cell lung cancer: first-, second-, and third-line.Curr. Oncol. 2012; 19: S52-S58Crossref PubMed Scopus (103) Google Scholar Approximately one-half of patients with advanced NSCLC never receive second-line therapy due to rapid disease deterioration during progression.18Lazzari C. Bulotta A. Ducceschi M. Viganò M.G. Brioschi E. Corti F. Gianni L. Gregorc V. Historical evolution of second-line therapy in non-small cell lung cancer.Front. Med. (Lausanne). 2017; 4: 4PubMed Google Scholar The trial from KEYNOTE-001 demonstrated pembrolizumab was associated with a median PFS of 12.5 months and a median OS of 22.1 months for treatment-naive patients and 10.6 months for previously treated patients (PD-L1 TPS ≥ 1%).19Hui R. Gandhi L. Costa E.C. Felip E. Ahn M.-J. Eder J.P. Balmanoukian A.S. Leighl N.B. Aggarwal C. Horn L. et al.Long-term OS for patients with advanced NSCLC enrolled in the KEYNOTE-001 study of pembrolizumab (pembro).J. Clin. Oncol. 2016; 34: 9026Crossref Google Scholar Compared with docetaxel, treatment with pembrolizumab was associated with prolonged OS in previously treated, PD-L1 TPS ≥ 1%, advanced NSCLC.7Herbst R.S. Baas P. Kim D.W. Felip E. Pérez-Gracia J.L. Han J.Y. Molina J. Kim J.H. Arvis C.D. Ahn M.J. et al.Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial.Lancet. 2016; 387: 1540-1550Abstract Full Text Full Text PDF PubMed Scopus (4549) Google Scholar These data confirmed pembrolizumab provides superior OS benefit for PD-L1-positive untreated and previously treated NSCLC. When administrated as first-line therapy, pembrolizumab provided significantly prolonged PFS and OS over chemotherapy for metastatic NSCLC with PD-L1 TPS ≥ 50%.20Brahmer J.R. Rodriguez-Abreu D. Robinson A.G. Hui R. Csõszi T. Fülöp A. Gottfried M. Peled N. Tafreshi A. Cuffe S. et al.Progression after the next line of therapy (PFS2) and updated OS among patients (pts) with advanced NSCLC and PD-L1 tumor proportion score (TPS) ≥50% enrolled in KEYNOTE-024.J. Clin. Oncol. 2017; 35: 9000Crossref Google Scholar, 21Reck M. Rodríguez-Abreu D. Robinson A.G. Hui R. Csőszi T. Fülöp A. Gottfried M. Peled N. Tafreshi A. Cuffe S. et al.KEYNOTE-024 InvestigatorsPembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer.N. Engl. J. Med. 2016; 375: 1823-1833Crossref PubMed Scopus (6215) Google Scholar Similarly, in the KEYNOTE-042 trial, patients with PD-L1 TPS ≥ 1%, the superiority of pembrolizumab over platinum-based chemotherapy is confirmed in treatment-naive metastatic NSCLC without epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) driver mutations.22Lopes G. Wu Y.-L. Kudaba I. Kowalski D. Cho B.C. Castro G. Srimuninnimit V. Bondarenko I. Kubota K. Lubiniecki G.M. et al.Pembrolizumab (pembro) versus platinum-based chemotherapy (chemo) as first-line therapy for advanced/metastatic NSCLC with a PD-L1 tumor proportion score (TPS) ≥ 1%: open-label, phase 3 KEYNOTE-042 study.J. Clin. Oncol. 2018; 36: LBA4Crossref Google Scholar These data provide strong rationale to extend pembrolizumab monotherapy as a standard first-line treatment for PD-L1-positive metastatic NSCLC. Chemotherapy stimulates anti-tumor immunity in two major ways: inducing immunogenic cell death (ICD)1Torre L.A. Siegel R.L. Ward E.M. Jemal A. Global cancer incidence and mortality rates and trends--an update.Cancer Epidemiol. Biomarkers Prev. 2016; 25: 16-27Crossref PubMed Scopus (2343) Google Scholar and disrupting the immune-suppressive tumor microenvironment that tumor uses to escape immune surveillance.2Devesa S.S. Bray F. Vizcaino A.P. Parkin D.M. International lung cancer trends by histologic type: male:female differences diminishing and adenocarcinoma rates rising.Int. J. Cancer. 2005; 117: 294-299Crossref PubMed Scopus (623) Google Scholar On cellular lever, the hallmark of ICD includes calreticulin translocation to cell membrane, which serves as a signal to dendritic cells (DCs) to clear the dying cell, and the release of ATP and high mobility group box protein 1 (HMGB1), which stimulate DC activation and maturation.23Kroemer G. Galluzzi L. Kepp O. Zitvogel L. Immunogenic cell death in cancer therapy.Annu. Rev. Immunol. 2013; 31: 51-72Crossref PubMed Scopus (1950) Google Scholar Treatment with oxaliplatin stimulates calreticulin exposure and HMGB1 release in colon cancer cells, and injection of oxaliplatin-treated cells into mice induced an anti-tumor immune response, which is mediated through HMGB1-TLR4 (Toll-like receptor) axis.24Tesniere A. Schlemmer F. Boige V. Kepp O. Martins I. Ghiringhelli F. Aymeric L. Michaud M. Apetoh L. Barault L. et al.Immunogenic death of colon cancer cells treated with oxaliplatin.Oncogene. 2010; 29: 482-491Crossref PubMed Scopus (763) Google Scholar Similarly, oxaliplatin-mafosfamide combination stimulated calreticulin exposure and HMGB1 release in lung adenocarcinoma models. Platinum-based chemotherapy modulates the composition and activity of tumor-infiltrating immune cells, which include increased infiltration of CD8+ T cells, maturation of antigen-presenting cells (APC), and downregulation of regulatory T cells (Tregs) and MDSCs at the tumor sites.25Liu W.M. Fowler D.W. Smith P. Dalgleish A.G. Pre-treatment with chemotherapy can enhance the antigenicity and immunogenicity of tumours by promoting adaptive immune responses.Br. J. Cancer. 2010; 102: 115-123Crossref PubMed Scopus (238) Google Scholar, 26Suzuki E. Kapoor V. Jassar A.S. Kaiser L.R. Albelda S.M. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity.Clin. Cancer Res. 2005; 11: 6713-6721Crossref PubMed Scopus (852) Google Scholar, 27Tseng C.-W. Hung C.-F. Alvarez R.D. Trimble C. Huh W.K. Kim D. Chuang C.M. Lin C.T. Tsai Y.C. He L. et al.Pretreatment with cisplatin enhances E7-specific CD8+ T-cell-mediated antitumor immunity induced by DNA vaccination.Clin. Cancer Res. 2008; 14: 3185-3192Crossref PubMed Scopus (131) Google Scholar Increased CD8+ T cell infiltration and CD8+ T cell:Treg ratio was also observed in oxaliplatin-mafosfamide-treated mice, which sensitize the tumor to immune checkpoint blockade.28Pfirschke C. Engblom C. Rickelt S. Cortez-Retamozo V. Garris C. Pucci F. Yamazaki T. Poirier-Colame V. Newton A. Redouane Y. et al.Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy.Immunity. 2016; 44: 343-354Abstract Full Text Full Text PDF PubMed Scopus (600) Google Scholar In addition, exposure to platinum chemotherapeutic drugs inhibit expression of programmed death-receptor-ligand 2 (PD-L2) on both tumor cells and dendritic cells (DCs), resulting in enhanced T cell stimulation and antigen reorganization.29Lesterhuis W.J. Punt C.J. Hato S.V. Eleveld-Trancikova D. Jansen B.J. Nierkens S. Schreibelt G. de Boer A. Van Herpen C.M. Kaanders J.H. et al.Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice.J. Clin. Invest. 2011; 121: 3100-3108Crossref PubMed Scopus (223) Google Scholar Other immune-stimulating effects of platinum-based drugs include increased major histocompatibility complex (MHC) class I expression and sensitizing tumor cells to granzyme-B-mediated cytotoxic T cell killing.12Ramakrishnan R. Assudani D. Nagaraj S. Hunter T. Cho H.I. Antonia S. Altiok S. Celis E. Gabrilovich D.I. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice.J. Clin. Invest. 2010; 120: 1111-1124Crossref PubMed Scopus (360) Google Scholar, 25Liu W.M. Fowler D.W. Smith P. Dalgleish A.G. Pre-treatment with chemotherapy can enhance the antigenicity and immunogenicity of tumours by promoting adaptive immune responses.Br. J. Cancer. 2010; 102: 115-123Crossref PubMed Scopus (238) Google Scholar Based on these findings, it is well acknowledged that platinum-based chemotherapy promotes the development of tumor-specific immunity and combining with immune checkpoint blockade may boost the function of effector T cells (Figure 1). Preclinical studies have confirmed that platinum-based chemotherapy elicits tumor CD8+ T cell infiltration and sensitized tumor to immune checkpoint blockade, which resulted in a durable control of cancer.28Pfirschke C. Engblom C. Rickelt S. Cortez-Retamozo V. Garris C. Pucci F. Yamazaki T. Poirier-Colame V. Newton A. Redouane Y. et al.Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy.Immunity. 2016; 44: 343-354Abstract Full Text Full Text PDF PubMed Scopus (600) Google Scholar More importantly, clinical studies have showed promising immunogenic effects of platinum. Compared to chemotherapy alone, combination therapy with ipilimumab and carboplatin and paclitaxel resulted in an improved PFS in a randomized phase II study in NSCLC in first-line setting.30Lynch T.J. Bondarenko I. Luft A. Serwatowski P. Barlesi F. Chacko R. Sebastian M. Neal J. Lu H. Cuillerot J.M. Reck M. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study.J. Clin. Oncol. 2012; 30: 2046-2054Crossref PubMed Scopus (848) Google Scholar A similar efficacy was also observed in a phase II trial of small-cell lung cancer.31Reck M. Bondarenko I. Luft A. Serwatowski P. Barlesi F. Chacko R. Sebastian M. Lu H. Cuillerot J.M. Lynch T.J. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial.Ann. Oncol. 2013; 24: 75-83Crossref PubMed Scopus (504) Google Scholar A randomized, controlled phase II trial has showed that addition of pembrolizumab to carboplatin and pemetrexed improved efficacy and resulted in a superior benefit-to-risk profile in chemotherapy-naive, metastatic NSCLC patients irrespective of PD-L1 expression, with a median PFS in the pembrolizumab combination group of 24 months compared to 9.3 months in the chemo alone group.32Langer C.J. Gadgeel S.M. Borghaei H. Papadimitrakopoulou V.A. Patnaik A. Powell S.F. Gentzler R.D. Martins R.G. Stevenson J.P. Jalal S.I. et al.KEYNOTE-021 investigatorsCarboplatin 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: 1497-1508Abstract Full Text Full Text PDF PubMed Scopus (1054) Google Scholar, 33Borghaei H. Langer C.J. Gadgeel S. Papadimitrakopoulou V.A. Patnaik A. Powell S.F. Gentzler R.D. Martins R.G. Stevenson J.P. Jalal S.I. et al.24-month overall survival from KEYNOTE-021 cohort G: pemetrexed and carboplatin with or without pembrolizumab as first-line therapy for advanced nonsquamous non-small cell lung cancer.J. Thorac. Oncol. 2019; 14: 124-129Abstract Full Text Full Text PDF PubMed Scopus (144) Google Scholar These findings were validated by the phase III KEYNOTE-189 trial in first-line setting: the triplet combination regimen significantly prolonged PFS and OS compared with cisplatin or carboplatin plus pemetrexed chemotherapy alone in metastatic nonsquamous NSCLC patients without sensitizing EGFR or ALK mutations.34Gandhi L. Rodríguez-Abreu D. Gadgeel S. Esteban E. Felip E. De Angelis F. Domine M. Clingan P. Hochmair M.J. Powell S.F. et al.KEYNOTE-189 InvestigatorsPembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer.N. Engl. J. Med. 2018; 378: 2078-2092Crossref PubMed Scopus (3533) Google Scholar The superior efficacy of this triplet regimen was also observed in a phase III trial of untreated metastatic, squamous NSCLC.35Paz-Ares L. Luft A. Vicente D. Tafreshi A. Gümüş M. Mazières J. Hermes B. Çay Şenler F. Csőszi T. Fülöp A. et al.KEYNOTE-407 InvestigatorsPembrolizumab plus chemotherapy for squamous non-small-cell lung cancer.N. Engl. J. Med. 2018; 379: 2040-2051Crossref PubMed Scopus (1995) Google Scholar Largely based on those trials, the Food and Drug Administration (FDA) approved the use of pembrolizumab along with pemetrexed and carboplatin as a first line of treatment for patients with metastatic, nonsquamous NSCLC in September 2017. More interestingly, the IMpower150 trial demonstrated that adding PD-L1 inhibitor atezolizumab to bevacizumab plus chemotherapy with carboplatin and paclitaxel in first line setting significantly improved PFS and OS among patients with advanced nonsquamous NSCLC, regardless of EGFR or ALK mutations and PD-L1 expression status.36Socinski M.A. Jotte R.M. Cappuzzo F. Orlandi F. Stroyakovskiy D. Nogami N. Rodríguez-Abreu D. Moro-Sibilot D. Thomas C.A. Barlesi F. et al.IMpower150 Study GroupAtezolizumab for first-line treatment of metastatic nonsquamous NSCLC.N. Engl. J. Med. 2018; 378: 2288-2301Crossref PubMed Scopus (2113) Google Scholar This suggests that blocking immunosuppression mediated by vascular endothelial growth factor may enhance the efficacy of immunotherapy. Several phase I and II clinical trials of chemotherapy combining with checkpoint blockade enrolling patients with different stages and histologies are currently ongoing (Table 1). These trials are investigating (1) whether combining checkpoint blockade with new chemotherapy drugs can help, (2) whether combining checkpoint blockade with standard chemotherapy is superior in adjuvant and neoadjuvant setting, (3) whether combining checkpoint blockade with standard chemotherapy is effective in earlier stages of disease, and (4) whether combining checkpoint blockade with other chemotherapy or targeting therapy drugs will overcome resistance to checkpoint inhibitor treatment. Their primary end points include safety and toxicity, maximum tolerated dose, objective response rate (ORR), PFS, OS, and alterations of immune response. Results from these trials will better establish the efficacy and toxicity of this regimen to better understand the immune evolution following combination treatment, to explore regimens that can overcome checkpoint inhibition resistance, and may extend this approach to earlier stages.Table 1Ongoing Studies of Immunotherapy in Combination with Chemotherapy or Targeting Therapy in Lung CancerNCT NumberStudy PhaseDisease StageTrial DesignLine of TherapyNCT03233724phase I and IIinoperable stage I–IV NSCLC (n = 75)tetrahydrouridine-decitabine (THU-DAC) with pembrolizumab1st lineNCT03823625phase IIstage IIIB not amenable to radical treatment or stage IV or recurrent squamous-cell lung carcinoma (SqLC) (n = 112)nivolumab plus ipilimumab versus platinum-based chemotherapy plus nivolumab1st lineNCT03801304phase IIstage IV NSCLCatezolizumab plus vinorelbine2nd lineNCT03568097phase IIstage IV SCLC (n = 55)avelumab + standard 1st line chemotherapy (cisplatin or carboplatin + etoposide)1st lineNCT02581943 (immune markers)phase IIrecurrent or stage IIIB–IV NSCLC (n = 40)pembrolizumab versus pembrolizumab + carboplatin + paclitaxel2nd and 3rd lineNCT03257722phase Ib and IINSCLC patients who have failed immune checkpoint inhibitor (n = 40)pembrolizumab + idelalisibany lineNCT03689855phase IIstage IV NSCLC patients progressed on any immune checkpoint blocker (n = 21)ramucirumab + atezolizumabany lineNCT02638090phase I and IIstage IV NSCLC (n = 100)pembrolizumab + vorinostatany line Open table in a new tab Various tissue- and plasma-based tests have been investigated with the aim of identifying biomarkers related to benefit from checkpoint blockade. A recent study using whole-exome sequencing demonstrated higher tumor mutation burden (TMB) was associated with improved objective response rate and prolonged PFS in NSCLC patients treated with pembrolizumab.37Rizvi N.A. Hellmann M.D. Snyder A. Kvistborg P. Makarov V. Havel J.J. Lee W. Yuan J. Wong P. Ho T.S. et al.Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.Science. 2015; 348: 124-128Crossref PubMed Scopus (5531) Google Scholar This observation was confirmed by Foundation One database analysis, where high TMB (>15 mutations/MB) was associated with longer clinical response in NSCLC patients treated with PD-1 or PD-L1 inhibitors.38Spigel D.R. Schrock A.B. Fabrizio D. Frampton G.M. Sun J. He J. Gowen K. Johnson M.L. Bauer T.M. Kalemkerian G.P. et al.Total mutation burden (TMB) in lung cancer (LC) and relationship with response to PD-1/PD-L1 targeted therapies.J. Clin. Oncol. 2016; 34: 9017Crossref Google Scholar In addition, POPLAR trial and Checkmate 026 trial showed similar observation, where higher TMB was associated with improved efficacy in NSCLC patients treated with PD-1 or PD-L1 inhibitor in the first- and second-line setting.39Kowanetz M. Zou W. Shames D. Cummings C. Rizvi N. Spira A. Frampton G. Leveque V. Flynn S. Mocci S. et al.OA20.01 Tumor mutation burden (TMB) is associated with improved efficacy of atezolizumab in 1L and 2L+ NSCLC patients.J. Thorac. Oncol. 2017; 12: S321-S322Abstract Full Text Full Text PDF Google Scholar, 40Peters S. Creelan B. Hellmann M.D. Socinski M.A. Reck M. Bhagavatheeswaran P. Chang H. Geese W.J. Paz-Ares L. Carbone D.P. Abstract CT082: impact of tumor mutation burden on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung cancer: an exploratory analysis of CheckMate 026.Cancer Res. 2017; 77: CT082Crossref Google Scholar Historically, PD-L1 expression has been used as a cutoff for selecting patients in numerous clinical trials. Although studies have demonstrated that high PD-L1 expression increases the likelihood of response to PD-1 blockade therapy, responses in tumors lacking PD-L1 expression was also reported. Interestingly, the heterogeneity of PD-L1 expression in tumor microenvironment (TME) may be a key factor affecting the efficacy of immunotherapy. PD-L1 expression on tumor-infiltrating immune cells (macrophages and dendritic cells), but not tumor cells, correlated with responses,41Herbst R.S. Soria J.C. Kowanetz M. Fine G.D. Hamid O. Gordon M.S. Sosman J.A. McDermott D.F. Powderly J.D. Gettinger S.N. et al.Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients.Nature. 2014; 515: 563-567Crossref PubMed Scopus (3695) Google Scholar which may explain the lack of efficacy of checkpoint blockade in a subset of PD-L1-high patients. High tumor-infiltrating lymphocyte (TIL) presence has been associated with superior prognosis in multiple solid tumors, including NSCLC.42Al-Shibli K.I. Donnem T. Al-Saad S. Persson M. Bremnes R.M. Busund L.T. Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer.Clin. Cancer Res. 2008; 14: 5220-5227Crossref PubMed Scopus (463) Google Scholar In a retrospective study of NSCLC patients treated with nivolumab, high CD8+PD-1low TILs in pre-treated samples was associated with prolonged PFS.43Mazzaschi G. Madeddu D. Falco A. Bocchialini G. Goldoni M. Sogni F. Armani G. Lagrasta C.A. Lorusso B. Mangiaracina C. 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. 2018; 24: 407-419Crossref PubMed Scopus (157) Google Scholar Interestingly, in a phase II trial of nivolumab for high-risk stage IB–IIIA NSCLC patients in neoadjuvant setting, comparing to pre-treatment, an influx of PD-1+CD8+ T cell infiltration in TME after treatment was associated with clinical response.44Chaft J.E. Forde P.M. Smith K.N. Anagnostou V. Cottrell T. Taube J.M. Rekhtman N. Merghoub T. Jones D.R. Hellmann M.D. et al.Neoadjuvant nivolumab in early-stage, resectable non-small cell lung cancers.J. Clin. Oncol. 2017; 35: 8508Crossref Google Scholar In a phase II trial of atezolizumab versus docetaxel for patients with previously treated NSCLC, increased PD-L1 expression on tumor cells and tumor-infiltrating immune cells and high T-effector (Teff)-associated gene expression was associated with improvement in OS.45Fehrenbacher L. Spira A. Ballinger M. Kowanetz M. Vansteenkiste J. Mazieres J. Park K. Smith D. Artal-Cortes A. Lewanski C. et al.POPLAR Study GroupAtezolizumab 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: 1837-1846Abstract Full Text Full Text PDF PubMed Scopus (1990) Google Scholar The Teff gene expression signature was further confirmed in a subsequent phase III trial, which showed high Teff expression identified more patients who are likely to experience a clinical benefit with atezolizumab than using PD-L1 expression as a biomarker.46Kowanetz M. Zou W. Mccleland M. Gandara D.R. Gadgeel S. Rittmeyer A. Barlesi F. Park K. Shames D. Koeppen H. et al.MA 05.09 Pre-existing immunity measured by Teff gene expression in tumor tissue is associated with atezolizumad efficacy in NSCLC.J. Thorac. Oncol. 2017; 12: S1817-S1818Abstract Full Text Full Text PDF Google Scholar In sum, multiple studies have suggested TMB as a strong biomarker for clinical benefit from PD-1 or PD-L1 inhibitors. The heterogeneity of PD-L1 expression and dynamic changes of CD8+ TILs and its interplay with other components of TME (such as Teff) during disease course needs more thorough study to establish a clinical cutoff for its prognostic and predictive value. Meanwhile, other assays that have demonstrated predictability of efficacy of immunotherapy in melanoma and other solid tumors, such as T cell clonality, immunoscore, immunophenoscore, and others, are under investigation in NSCLC. Despite the unparalleled durable response with checkpoint inhibitors, the majority of patients do not benefit (primary resistance) and some patients relapse after an initial response (acquired resistance). To this point, most studies of mechanisms of resistance to immunotherapy were performed in melanoma, which includes tumor cell intrinsic and extrinsic factors. The tumor-cell-intrinsic factors include low neoantigen, deficient antigen presentation, insensitivity to interferon gamma pathway signaling, as well as genetic T cell exclusion, such as overactivation of wnt and β-catenin signaling. Tumor-cell-extrinsic factors include upregulation of alternative inhibitory checkpoint molecules; infiltration of other immunosuppressive cell populations, such as macrophages and regulatory T cells; as well as lack of specific anti-tumor T cell responses.47Sharma P. Hu-Lieskovan S. Wargo J.A. Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy.Cell. 2017; 168: 707-723Abstract Full Text Full Text PDF PubMed Scopus (2485) Google Scholar In NSCLC, trials have shown that patients with EGFR mutant tumors do not benefit from PD-1 or PD-L1 inhibitors.48Rittmeyer A. Barlesi F. Waterkamp D. Park K. Ciardiello F. von Pawel J. Gadgeel S.M. Hida T. Kowalski D.M. Dols M.C. et al.OAK Study GroupAtezolizumab 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: 255-265Abstract Full Text Full Text PDF PubMed Scopus (3166) Google Scholar, 49Borghaei H. Paz-Ares L. Horn L. Spigel D.R. Steins M. Ready N.E. Chow L.Q. Vokes E.E. Felip E. Holgado E. et al.Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer.N. Engl. J. Med. 2015; 373: 1627-1639Crossref PubMed Scopus (6773) Google Scholar Further studies have demonstrated that EGFR mutant and ALK-rearrangement-positive NSCLC tumors lacked concurrent CD8+ T cell infiltration and PD-L1 expression, which is associated with inferior efficacy of PD-1 or PD-L1 inhibitors in this subset of the population.50Gainor J.F. Shaw A.T. Sequist L.V. Fu X. Azzoli C.G. Piotrowska Z. Huynh T.G. Zhao L. Fulton L. Schultz K.R. 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. 2016; 22: 4585-4593Crossref PubMed Scopus (816) Google Scholar Quite contrary to this observation, a recent phase III trial showed significant efficacy in this subset of patients treated with atezolizumab and bevacizumab plus chemotherapy with carboplatin and paclitaxel.36Socinski M.A. Jotte R.M. Cappuzzo F. Orlandi F. Stroyakovskiy D. Nogami N. Rodríguez-Abreu D. Moro-Sibilot D. Thomas C.A. Barlesi F. et al.IMpower150 Study GroupAtezolizumab for first-line treatment of metastatic nonsquamous NSCLC.N. Engl. J. Med. 2018; 378: 2288-2301Crossref PubMed Scopus (2113) Google Scholar Another phenomenon worth noting is hyperprogressive (HP) disease in patients treated with immunotherapy, which occurs in about 17% metastatic NSCLC patients after starting checkpoint inhibitors.51Kim Y. Kim C.H. Kim H.S. Sun J.-M. Ahn J.S. Ahn M.-J. Lee S.-H. Lee H.Y. Park K. Hyperprogression after immunotherapy: clinical implication and genomic alterations in advanced non-small cell lung cancer patients (NSCLC).J. Clin. Oncol. 2018; 36: 9075Crossref Google Scholar One possible explanation is accumulation of M2-like macrophages in pre-treatment tumors, and the interaction between Fc-Fcγ receptor binding of Nivolumab and macrophages facilitated tumor progression.52Lo Russo G. Moro M. Sommariva M. Cancila V. Boeri M. Centonze G. Ferro S. Ganzinelli M. Gasparini P. Huber V. et al.Antibody-Fc/FcR interaction on macrophages as a mechanism for hyperprogressive disease in non-small cell lung cancer subsequent to PD-1/PD-L1 blockade.Clin. Cancer Res. 2018; (Published online December 17, 2018)https://doi.org/10.1158/1078-0432.CCR-18-1390Crossref PubMed Scopus (251) Google Scholar Further investigations are warranted to interrogate the mechanisms underlying resistance and hyperprogressive disease of checkpoint inhibitors. Platinum-based chemotherapy has been the treatment of choice of advanced NSCLC for years in the first-line setting. The development of checkpoint inhibitors targeting PD-1 or PD-L1 pathway has drastically changed the treatment paradigm of metastatic NSCLC. The extensive clinical investigations of combination of pembrolizumab with carboplatin and pemetrexed have facilitated the FDA approval of the first combination of chemotherapy and immunotherapy for patients with lung cancer. Despite having more therapeutic opportunities, this has also increased the complexity of therapeutic selection. In the age of precise medicine, the most urgent challenge that remains is the lack of biomarkers to predict which patient may benefit the most from combination of chemotherapy and checkpoint blockade and which patient may experience hyperprogressive tumor progression. Growing evidence suggests that the genomic context of the tumor may have a critical influence on the tumor microenvironment, thus impacting the efficacy of immunotherapy. This can be partially interrogated via next-generation sequencing where the tumor genetics and efficacy of selected treatment can be studied. In addition, the dissection of immune infiltration of TME via high-resolution multiplex technique, such as CyTOF, can provide a more fundamental understanding of the immune component of TME and its association with treatment selection and efficacy. Another increasing concern is the acquired resistance to therapy after initial tumor regression. Additional investigations on elucidating the mechanisms underlying acquired mechanism(s) that are currently underway may help in developing combination strategies that benefit patient in the long term.