Abstract
Advances in the understanding of the role of the immune system in tumor immunosurveillance have resulted in the recognition that tumors can evade immune destruction via the dysregulation of co-inhibitory or checkpoint signals. This has led to the development of a generation immunotherapeutic agents targeting the immune checkpoint pathway. Recent early phase studies of immune checkpoint modulators, such as CTLA-4, PD-1 and PD-L1 inhibitors in NSCLC have reported promising results with prolonged clinical responses and tolerable toxicity. This article provides an overview of co-stimulatory and inhibitory molecules that regulate the immune response to tumors, recent therapies that have been developed to exploit these interactions and the role of predictive biomarkers in treatment selection.
Keywords: Immunotherapy, Non-small cell lung cancer, PD-L1, CTLA-4, Ipilimumab, Nivolumab
1. Background
Lung cancer is one of the most common malignancies in the world, with 1.6 million new cases diagnosed annually and is also the leading cause of cancer deaths worldwide, causing 1.4 million deaths annually [1]. Whilst the identification of activating sensitizing EGFR mutations and other driver oncogenes [2,3] has led to improved treatment outcomes in selected subgroups of patients with advanced stage NSCLC, survival remains dismal and novel therapeutic approaches are needed [4].
Tumorigenesis is not only dependent on the properties of cancer cells but also by interaction with the immune system [5]. Testament to this is the numerous immunotherapies in clinical use in cancer, such as Sipuleucel-T (metastatic castrate resistant prostate cancer), ipilimumab and interleukin (IL)-2 (advanced melanoma) and IL-2 (renal cell carcinoma). Nonetheless, NSCLC has traditionally been thought to be a non-immunogenic tumor due to the inactivity of similar agents such as Bacillus Calmette-Guerin (BCG) [6], IL-2 [4] and interferon [7] have been unsuccessful in NSCLC. However the development new generation of cancer vaccines and immune modulators has renewed interest in immunotherapy in NSCLC. With respect to vaccines, although initial studies were promising, several phase III trials have been disappointing. Studies with tecemotide (START study) [8], and belagenpumatucel-L (STOP study) [9] in the locally advanced setting, and melanoma associated antigen A3 (MAGE-A3) (MAGRIT study) in the adjuvant setting [10] have been negative. The role of cancer vaccines has been reviewed elsewhere and will not be discussed [11].
The expression of antigens in cancer cells differs from host cells due to genetic and epigenetic variations. In order for the immune system to eliminate cancer cells, tumor recognition must first occur. This is followed by tumor antigen presentation to T cells, which leads to T cell activation and finally cell kill can occur. Co-stimulatory and inhibitory signals regulate T cell mediated immune response. T cell mediated immune response is modulated by stimulatory and inhibitory signals. Immune co-stimulatory molecules include CD28, CD137, glucocorticoid-induced tumor necrosis factor (TNF) receptor (GITR), and OX-40. Negative regulatory molecules immune checkpoint molecules prevent overstimulation of immune responses. Checkpoint molecules (co-inhibitory molecules) include cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), T-cell immunoglobulin- and mucin domain-3-containing molecule 3 (TIM3), Lymphocyte-activation gene 3 (LAG3) and killer cell immunoglobulin-like receptor (KIR). (Table 1) These immune checkpoints exist in a normal physiological state to protect against autoimmunity and inflammation. In a neoplastic state, dysfunction of these immune checkpoint proteins can lead to tumor tolerance and eventually allow for tumor “escape” from the immune system.
Table 1.
Co-inhibitory and co-stimulatory molecules and their binding partners.
| Receptor
|
Binding partner
|
|||||
|---|---|---|---|---|---|---|
| Name | Expression in immune cells | Therapeutic agent | Name | Expression in normal cells | Therapeutic agent | References |
| Co-inhibitory molecules | ||||||
| CTLA-4 | Activated T cells, Tregs | Ipilimumab, Tremelimumab | CD80, CD86 | Activated B cells, monocytes | [12] | |
| PD-1 | Activated CD4, CD8, Tregs, Activated B cells and NK cells | Nivolumab, Lambrolizumab, pidilizumab (CT-011), AMP-224 | PD-L1(B7-H1), PD-L2 | Pancreas, thymus, liver, heart, endothelium, small intestine | PD-L1: MDX-1105, MEDI4736, MPDL3280A | [13,14] |
| TIM3 | Monocytes, NK cells, CD4TH1 cells | Galectin-9 | Small intestine | [15,16] | ||
| LAG3 (CD223) | CD4, CD8, γδ T cells, Treg, B cells, NK cells and plasmacytoid DC | IMP321 | MHC-II | Antigen presenting cells, B lymphocytes | [17] | |
| BTLA | TH1 | HVEM | Peripheral T and B cells | [18,19] | ||
| A2aR | Ubiquitous | Adenosine B7-H3.B7-H4 | Ubiquitous Haematopoetic cells |
MGA271 | [20] [21] |
|
| KIR | NK cells | Lirilumab | MHC-II or MHC-I | Antigen presenting cells, B lymphocytes | [22] | |
| Co-stimulatory molecules | ||||||
| OX-40(CD134) | Activated T cells, Treg | MEDI6469 | OX40-L | Peripheral B lymphocytes | R04989991 | [23] |
| CD137(4-1BB) | Activated T cells, dendritic cells, NK cells | BMS-666513, PF-05082566 | CD137L | Most leukocytes | [24] | |
| CD40 | Dendritic cells, B lymphocytes, macrophages, endothelial cells, smooth muscle cells, fibroblasts, monocytes | Dacetuzumab, Lucatumumab, CP-870893 | CD40L(CD154) | CD4, CD8, skin mast cells and blood basophils | [25,26] | |
| CD27 | Resting T lymphocytes | CDX1127 | CD70 | Activated T and B lymphocytes | [27] | |
| GITR | Treg, CD4, CD8, | TRX518 | GITR-L | Antigen presenting cells | [28] | |
| ICOS | Activated T cells | MEDI570, AMG557 | B7RP1 | B cells, macrophages | [29] | |
| CD28 | T cells, eosinophils | TGN1412 | CD80, CD86 | Activated B cells, monocytes | [30] | |
A2aR: A2A adenosine receptor; BTLA: B and T lymphocyte attenuator; DC: dendritic cells; GITR: glucocorticoid-induced tumor necrosis factor receptor; HVEM: herpes virus entry mediator; ICOS: Inducible T-cell co-stimulator; KIR: killer cell immunoglobulin-like receptor; LAG3: lymphocyte-activation gene 3; PD-1: programmed death-1; NK: natural killer; PD-L1: programmed death receptor ligand 1; TIM3: T-cell immunoglobulin- and mucin domain-3-containing molecule 3; Tregs: T regulatory cells.
Targeting the molecules that regulate the immune response using antibodies has been the subject of much research and has yielded some promising results. This article provides an overview of co-stimulatory and inhibitory molecules that regulate the immune response to tumors, and recent therapies that have been developed to exploit these interactions.
2. Co-inhibitory interactions
2.1. CTLA4
The presentation of an antigen to T cell receptor (TCR) by a major histocompatibility complex on an antigen presenting cell (APC) as well as the binding of B7 molecules on APCs with the CD28 receptors on the T cells leads to the activation of CD4 and CD8 T cells. B7 has two subtypes of ligands, B7.1 or CD80 and B7.2 or CD86. Cytotoxic T lymphocyte antigen-4 (CTLA-4) is also binds to B7, and acts competitively with CD28, but in an inhibitory fashion. The CTLA4 binds to B7, it releases signals that revert an activated T cell into an inhibited T cell. CTLA4 is up regulated only following T cell activation, and is only barely detectable on naïve T cells [31]. CTLA4 also reduces IL2 production; IL2 receptor expression and can directly inhibit TCR signals [32]. As a result of CTLA4 activation, peripheral tolerance of antigen specific T cells occurs. Mice with CTLA4-knockout have been shown to have phenomenal and lethal lymphoproliferation, indicating the importance of the role CTLA4 plays in inhibiting T cell activation [33]. CTLA4 expression is constitutive on regulatory T cells (Tregs) and is induced in CD4 and CD8 T cells.
CTLA-4 expression is not only confined to T lymphocytes but it is also expressed in NSCLC tumors in 51–87% of cases (Table 2). In one study CTLA-4 was associated with non-squamous histology but was not prognostic for overall survival [34] whilst in a second study CTLA-4 tumor expression was associated with older age and poorer tumor differentiation [35].
Table 2.
Tumor expression of CTLA-4, PD-L1 in resected NSCLC.
| Author | N | Histologic subtype | Pathologic stage | Detection method/Ab clone | Cellular localization | % PD- L1 + ve |
Clinpathological association |
Association with immune cells/TILs |
Prognosis |
|---|---|---|---|---|---|---|---|---|---|
| CTLA-4 | |||||||||
| Salvi [34] | 81 | Mixed | I–III | IHC anti CTLA-4/14D3 | Cell surface, cytoplasm | 50.6 | Non-squamous | Not reported | OS: neutral 5 year survival 64.8 vs. 45.9% |
| Zheng [35] | 89 | Mixed | I–IV | IHC anti-CTLA-4 | Not reported | 86.5 | Older age, poorer differentiation | Not reported | Not reported |
| PD-L1 | |||||||||
| Yang [45] | 163 | ADC | I | IHC anti-PD-L1/Proteintech Group Chicago, IL | Membrane | 39.9 | Vascular invasion, higher grade differentiation | No association with TILs | RFS: improved, OS: neutral |
| Velchetti [46] | 204 (US) | Mixed | I–IV | QIF/5H1 | Membrane | 36.1 | SCC | Increased inflammatory infiltrate | OS: improved 60 v 27months |
| 340 (Greece) | Mixed | I–IV | QIF/5H1 | Membrane | 24.8 | Lower stage | Increased inflammatory infiltrate | OS: improved NR v 31months | |
| 173 (US) | Mixed | I–IV | mRNA | Not applicable | 50.8 | None | Increased inflammatory infiltrate | OS: improved | |
| 314 (Greece) | Mixed | I–IV | mRNA | Not applicable | 53.2 | None | Increased inflammatory infiltrate | OS: improved | |
| Chen [47] | 208 | Mixed | I–IV | IHC anti-PD-L1 | Cytoplasm, membrane | 65.3 | Non-smokers, less LN metastasis | Increased macrophages | Not reported |
| Velchetti [52] | 445 | Mixed | I–IV | QIF/5H1 | Not reported | 27.4 | Not reported | Not reported | Not reported |
| 13 | Sarcomatoid | I–IV | QIF/5H1 | Not reported | 69.2 | Not reported | Not reported | Not reported | |
| Chen [48] | 120 | Mixed | I–III | IHC anti-PD-L1/236A/E7 | Cytoplasm, membrane | 57.5 | Not reported | Not reported | OS: reduced |
| Boland [49] | 214 | SCC | I–IV | IHC anti PD-L1/5H1 | Membrane | 19.6 | Not reported | No association with TILs | OS: neutral |
| Mu [50] | 109 | Mixed | I–III | IHC anti-PD-L1/not reported | Cytoplasm, membrane | 53.2 | ADC | Increased dendritic cells | OS: reduced PD-L1 + <3 year survival 46%, >3Y survival 12% |
| Konishi [51] | 52 | Mixed | I–IV | IHC anti-PD-L1/M1H1 | Cytoplasm, membrane | 27.2 | None | Reduced TILs | OS: neutral 5 year survival 59% v 48% |
Ab: antibody; ADC: adenocarcinoma; IHC: immunohistochemistry; LN: lymph node; NR: not reached; OS: overall survival; PD-L1: programmed death-1 ligand; QIF: quantitative fluorescence; RFS: relapsed free survival; SCC: squamous cell carcinoma; TILs: tumor infiltrating lymphocytes.
The anti-tumor effect of CTLA4 inhibition is via the reduction in the inhibition of CD28/B7 T cell activation. This also results in reduced Tregs, ultimately leading to an accelerated immune response to tumor associated antigens. The CTLA-4 inhibitor, ipilimumab has been shown to be effective in advanced stage melanoma and is currently being studied in NSCLC.
Ipilimumab is a humanized monoclonal antibody that binds to CTLA4, thereby preventing it from binding to its ligand and reducing the inhibition of CD28/B7 T cell activation by CTLA4. CTLA4 inhibition also reduces Tregs, ultimately leading to an accelerated immune response to tumor associated antigens.
In a randomized phase II trial in patients with advanced NSCLC [36], patients were randomized to receive one of the three regimens. All treatment arms received up to six cycles of paclitaxel and carboplatin. The first arm (early) received ipilimumab on D1 of cycle 1–4, and placebo for the remaining 2 cycles. The second arm received placebo for the first 2 cycles and ipilimumab on D1 of cycle 3–6 (delayed). The third arm (control) received only placebo. Maintenance ipilimumab was given in patients in the first two treatment groups once every 12 weeks until progression. Immune-related progression free survival (irPFS), which takes into account the immune related response criteria (described later in the article), was the study’s primary endpoint. In the delayed arm, the irPFS was 5.7 vs 4.6 months (HR = 0.72; P = 0.05), whilst in the early arm, no improvement in irPFS was seen (5.5 vs 4.6 months HR = 0.81; P = 0.13). In the delayed group, a non-statistical improvement in OS was also seen (12.2 vs 8.3 months HR = 0.87; P = 0.23). Although not statistically significant, patients with squamous histology had longer OS (HR = 0.55, 95% CI, 0.27–1.12).
The side effects reported were rash, pruritus and diarrhea. Grade 3/4 irAE was 20% for the early phase, 15% for the delayed phase and 6% for the control group. One death from toxic epidermal necrolysis was attributed to ipilimumab.
A larger phase III trial is being conducted, aiming specifically at the squamous subtype NSCLC (NCT01285609). Ipilimumab is also being studied in combination with EGFR and ALK tyrosine kinase inhibitors (NCT01998126). The role of ipilimumab is also being investigated in small cell lung cancer (NCT01331525, NCT01450761, NCT02046733).
Tremelimumab, a monoclonal antibody similar to ipilimumab has been studied in a phase II study of pre-treated patients with advanced stage NSCLC [37]. Patients were randomized into two arms-tremelimumab or best supportive case after 4 cycles of a platinum doublet chemotherapy regimen of investigators choice. The ORR was 5% and there was no difference in PFS.
2.2. PD1
PD-1 receptor is expressed on CD4 and CD8 lymphocytes, Tregs, B lymphocytes and NK cells [13]. Known ligands of PD-1 include PD-L1 (or CD274, B7-H1) and PD-L2 (CD 273, B7-DC). The binding of PD-1 with PD-L1 or PD-L2 leads to decreased cytokine production, reduced proliferation and cell lysis. In many tumors, PD-1 is up regulated in tumor infiltrating lymphocytes (TILs), while many tumors have increased PD-L1 expression [38]. It is proposed that through this mechanism, tumors can induce T cell anergy and avoid the processing tumor antigens by APCs that lead to recognition. PD-1 antagonists include PD-L1 antibodies such as nivolumab (BMS936558), lambrolizumab (MK-3475), and pidilizumab (CT-011) and the fusion protein AMP-224.
Nivolumab (BMS-936558, MDX-1106, ONO-4538) is a fully human IgG4 monoclonal antibody without detectable antibody-dependent cellular cytotoxicity (ADCC). In a phase I study of patients with advanced stage solid tumors [39], escalating doses of nivolumab biweekly were given for up to 12 cycles (2 years). In the NSCLC cohort (n = 129) the majority of patients were heavily pretreated, with 55% receiving at least 3 prior lines of therapy. The ORR was 17% with a median duration of response of 74 weeks (range, 6.1–133.9 weeks). The median survival was 9.9 months with one and two year survival rates of 42 and 24%, respectively. The median PFS was only 2.3 months. Nivolumab was generally well tolerated with skin toxicities (20%), gastrointestinal (15%) and pulmonary (9%) being the most commonly observed adverse events (AEs). A lower frequency of gastrointestinal toxicities was seen: 2% (grade 3/4) as compared to 20% with ipilimumab. Pneumonitis was reported in 6% (8/129) of patients with two deaths [40]. Biomarker analysis for PD-L1 expression was performed in 49% (63/129) patients. PD-L1 positive cases, defined as expression in at least 5% of tumor cells on immunohistochemistry (IHC), were seen in 49% (31/63) of patients. The ORR in patients with PD-L1 positive and PD-L1 negative tumors was 16% and 13%, respectively [41], suggesting that in a pretreated group, archival tumor tissue may not be ideal for assessing PD-L1 status.
Phase III trials of nivolumab versus docetaxel in patients with either squamous NSCLC (NCT01642004) or non-squamous NSCLC (NCT01673867) have completed accrual and results are eagerly awaited (Table 3).
Table 3.
Selected ongoing studies of immune checkpoint mediators.
| Phase | Patient population | N | Regimen | Control | Primary endpoint | Clinical trials.gov |
|---|---|---|---|---|---|---|
| CTLA-4 | ||||||
| I | ALK/EGFR mutant NSCLC 1st line | 46 | Ipilimumab + erlotinib Ipilimumab + crizotinib |
None | Toxicity | NCT01998126 |
| II | NSCLC 1st line | 204 | Ipilimumab + Carboplatin/paclitaxel Concurrent vs sequential |
Carboplatin/paclitaxel | irPFS | NCT00527735 |
| III | Squamous NSCLC 1st line | 920 | Ipilimumab + Carboplatin/paclitaxel | Carboplatin/paclitaxel | OS | NCT01285609 |
| I | EGFR mutant NSCLC 1st line | 24 | Tremelimumab + Gefitinib | None | Toxicity | NCT02040064 |
| II | NSCLC after 1st line, maintenance | 87 | Tremelimumab | BSC | PFS | NCT00312975 |
| PD-1 | ||||||
| I | EGFR mutant NSCLC 1st line | 24 | Tremelimumab + Gefitinib | None | Safety, RP2D | NCT02040064 |
| I | 1st line, Squamous, non-squamous NSCLC | 250 | Nivolumab in combination with platinum doublet, bevacizumab maintenance, erlotinib, ipilimumab, or nivolumab alone | None | Toxicity | NCT01454102 |
| II | Squamous NSCLC, 3rd or more line | 100 | Nivolumab | None | ORR | NCT01721759 |
| III | Nonsquamous NSCLC, 2nd line | 574 | Nivolumab | Docetaxel | OS | NCT01673867 |
| III | Squamous NSCLC, 2nd line | 264 | Nivolumab | Docetaxel | ORR | NCT01642004 |
| I | NSCLC 1st line | 30 | Lambrolizumab (MK-3475) monotherapy or in combination with platinum doublet | None | Toxicity | NCT01840579 |
| I | Pre-treated NSCLC, PD-L1+ | 24 | Lambrolizumab | None | ORR | NCT02007070 |
| I | Advanced stage melanoma/NSCLC | 1137 | Various doses of Lambrolizumab | None | Toxicity | NCT01295827 |
| I/II | NSCLC, any line | 320 | In combination with platinum doublet ± bevacizumab, erlotinib, gefitinib, or ipilimumab | None | PFS/ORR | NCT02039674 |
| II/III | NSCLC 2nd line after platinum doublet | 920 | Lambrolizumab low dose/high dose | Docetaxel | OS | NCT01905657 |
| PD-L1 | ||||||
| I | Advanced stage solid tumors | 20 | MSB0010718C | None | DLT | NCT01943461 |
| I | Advanced stage selected solid tumors | 590 | MSB0010718C | None | DLT | NCT01772004 |
| IB | EGFR mutant NSCLC 1st line | 32 | MPDL3280A + Erlotinib | None | DLT | NCT02013219 |
| II | PD-L1 + NSCLC, any line | 130 | MPDL3280A | None | ORR | NCT01846416 |
| II | PD-L1 + NSCLC, any line | 300 | MPDL3280A | None | ORR | NCT02031458 |
| II | NSCLC, 2nd line after platinum doublet | 300 | MPDL3280A | Docetaxel | OS | NCT01903993 |
| III | NSCLC, 2nd line after platinum doublet | 850 | MPDL3280A | Docetaxel | OS | NCT02008227 |
| IB | NSCLC, any line | 208 | MEDI4736 +tremelimumab | None | DLT, MTD | NCT02000947 |
| LAG3 | ||||||
| I | Advanced stage solid tumors | BMS-986016± nivolumab | None | Safety, DLT | NCT01968109 | |
| KIR | ||||||
| I | Advanced stage solid tumors | 150 | Lirilumab + nivolumab | None | DLT, MTD | NCT01714739 |
| I | Advanced stage solid tumors | 125 | Lirilumab + ipilimumab | None | DLT, MTD | NCT01750580 |
| CD27 | ||||||
| I | Advanced solid tumors | 170 | CDX-1127 | None | Toxicity | NCT01460134 |
ALK: anaplastic lymphoma kinase; BSC: best supportive care; DLT: dose limiting toxicity; EGFR: epidermal growth factor receptor; irPFS: immune related progression free survival; MTD: maximally tolerated dose; NSCLC: non-small cell lung cancer, ORR: overall response rate; OS: overall survival; PFS: progression free survival; RP2D: recommended phase II dose.
Lambrolizumab (MK-3475) is a monoclonal antibody targeting PD-1 with significant antitumor activity in melanoma [42]. Preliminary results from a NSCLC phase 1 expansion cohort, a median survival of 51 weeks and a partial response of 25% as assessed by immune related response criteria [43]. Common AEs were fatigue, rash and pruritis, whilst grade 2 pneumonitis (n = 1) and grade 3 pulmonary edema (n = 1) were reported. In the tumor biomarker studies, fresh pre-treatment tumor biopsies were obtained. Tumor PD-L1 expression by IHC was a predictor of response with the ORR of 67% (6/9) and 4% (1/24) in PD-L1 positive and negative tumors, respectively. Based on these results, a randomized phase II/III trial of lambrolizumab vs docetaxel in patients with PDL1 positive advanced NSCLC is being conducted (NCT01905657) (Table 3).
2.3. PDL1
Programmed death receptor ligand 1 (PD-L1, B7-H1), the ligand for PD-1, is a member of the B7 superfamily, is involved in the negative regulation of immune response [44]. PD-L1 is expressed in T and B cells, macrophages and dendritic cells and is up regulated in a range of solid tumors including NSCLC. Upon induction by cytokines such as interleukin-4 (IL-4), IL-10, interferon-α, -B or -γ, PD-L1 activates PD-1 on T cells, and down-regulates T cell effector function and is one mechanism by which cancer cells evade host immune surveillance. PD-L1 is expressed in 27–57.5% of NSCLC (Table 2) and is localized in the cell membrane ± cytoplasm [45–51]. In sarcomatoid NSCLC, PD-L1 is expressed in 69% of cases compared with 27% in NSCLC [52]. The prognostic role of PD-L1 is currently unclear with studies reporting no association [45,49,51], a worse overall survival [48,50] or improved survival [46]. The conflicting results could be attributed to different methodologies to detect and score PD-L1 expression. PD-L1 expression has been reported to be associated with vascular invasion and higher-grade differentiation but improved relapse free survival [45]. PD-L1 expression is associated with increased macrophages [47], increased dendritic cells [50] and increased inflammatory infiltrate [46]. In contrast, one study found PD-L1 expression was inversely related to TILs [51]. With regards to histology, PD-L1 expression was associated with squamous cell carcinoma [46] whilst in contrast, with adenocarcinoma in a separate study [50]. One study found no association between PD-L1 expression and EGFR/KRAS mutations or ALK rearrangement [45] (Table 2). Given the key role PD-L1 has in lung cancer, the inhibition of PD-L1 is an attractive therapeutic approach. PD-L1 inhibitors undergoing clinical development include the monoclonal antibodies MPDL3280A, BMS-936559, MEDI4736 and MSB0010718C (Table 3).
In the expanded phase I study of BMS-936559 (MDX-1105) in solid tumors, the ORR in NSCLC was 10% and 18% had stable disease (SD) of at least 24 weeks [53]. The development of BMS-936559, however, has apparently been halted.
MPDL3280A is an engineered IgG anti-PD-L1 antibody with modified Fc domain that prevents antibody-dependent cell-mediated cytotoxicity (ADCC) in other immune cells expressing PD-L1. In a phase I study of MPDL3280A in pre-treated patients with advanced NSCLC, the ORR was 24% and the 24-week PFS was 46%. The ORR in patients with PD-L1 positive and negative tumors was 100% (4/4) and 15% (4/26), respectively. Interestingly, the ORR in former/current smokers was 25% (8/31) versus 16% (1/6) in the never smokers [54].
MEDI4736, similar to MPDL3280A, is engineered with a triple mutation in the Fc domain to avoid Fc-mediated ADCC. Preliminary results from the phase I study of patients with solid tumors reported clinical activity and durable disease stabilization in different tumor types including NSCLC with no dose limiting toxicities or grade 3–4 treatment-related adverse events [55].
2.4. TIM3
Unlike other immune checkpoint molecules, TIM3 is not up regulated on all T cells post-activation but only in CD4+ T helper 1 (Th1) and CD8+ T cytotoxic cells and is involved in co-inhibition [56]). After activation by its ligand galectin-9, TIM3 inhibits effector Th1 cells and induces peripheral tolerance [57,58]. TIM3 plays a key role in the T cell exhaustion in tumors [59,60]. It can be co-expressed with PD-1 and in patients with melanoma; this co-expression represents the most exhausted population of CD8+ T cells in tumors [60]. In patients with NSCLC, TIM-3 in TILs is up regulated in CD4+ and CD8+ TILs but not expressed in peripheral blood T cells. The expression of TIM-3 in CD4+ T cells was associated with nodal metastasis and advanced cancer stage [61]. As well as being present in T cells, TIM3 has been reported in NSCLC in 86.7% of NSCLC tumor cells and is associated with T stage and histology and is an independent prognostic factor in NSCLC (relative risk of 4.481; 95% CI, 1.790–11.22) [62]. In preclinical models, the combined inhibition of TIM3 and PD-1 has been reported to be more effective than inhibiting either pathway alone in a range of solid tumors [59,63,64]. Taken together, the data supports the development of TIM3 inhibitors in humans.
2.5. LAG3
Lymphocyte-activation gene 3 (LAG3; CD223) is a co-inhibitory receptor expressed in activated T cells, Treg, Dendritic cells and NK cells. LAG3 is a CD4-related protein that binds to major histocompatibility complex (MHC) class II and reduces T cell proliferation resulting in tumor evasion [65,66]. LAG3 gene expression is up regulated in a silica-mediated lung inflammation murine model [67]. A phase I study of BMS-986016, a LAG3 monoclonal antibody with or without nivolumab in advanced solid tumors is currently ongoing (NCT01968109) (Table 3).
2.6. KIR
KIR is a family of natural killer (NK) cell regulators which presents a novel class of immunotherapy targets that are beginning to garner interest in various tumor types. KIR is one of the negative regulators of NK cell effector function. The inhibition of KIR results in NK cell mediated anti-tumor activity [68]. Lirilumab is an antibody that binds to KIR2DL1, −2 and −3 receptors, causing NK cell mediated cell kill [69]. Increased co-expression of KIR2DL1 in NSCLC patients has been described, which could potentially lead to decreased natural killer cell function [70]. Two phase I studies of lirilumab in combination with ipilimumab and nivolumab in patients with NSCLC are currently ongoing (NCT01714739 and NCT01750580) (Table 3).
2.7. BTLA and A2AR
Co-inhibitory receptor B and T lymphocyte attenuator (BTLA) has been studied in the role of autoimmunity, and along with its ligand herpes virus entry mediator (HVEM) has been associated with lymphomagenesis [71]. A2A adenosine receptor (A2AR) is a G protein-coupled receptor, which binds to adenosine and may potentially regulate the MAPKinase pathway [20]. Currently no studies are being conducted on these molecules in lung cancer.
3. Co-stimulatory interactions
3.1. OX40
OX-40 (CD134, TNFRSF4 tumor necrosis factor receptor superfamily, member 4) is a co-stimulatory molecule present in activated T cells at sites of inflammation [72] and regulates antigen-specific T-cell expansion, survival and cytokine IL-2, IL-4, IL-5, IFN-γ production [73,74]. The immunological effects of OX40 signaling make it an attractive target for improving tumor immunotherapy [75]. Pre-clinical studies have shown OX40 agonists have anti-tumor activity in melanoma, glioma, breast and colon carcinoma, sarcoma, renal carcinoma, and prostate cancer [76–79]. In a phase I study, patients with solid tumors were treated with one cycle of a murine agonistic anti-human OX40 Ab at three dose levels. Toxicities were acceptable and included fatigue, fever/chills, transient lymphopenia, and mild rash. Tumor reduction was seen in 12/30 patients and both humoral and cellular immunity were enhanced [80]. High levels of human anti-mouse antibodies were detected, thus future studies of humanized OX40 agonists are required for further development of this antibody [81]. Studies of OX40 agonist combined with radiotherapy are ongoing in breast and prostate cancer (NCT01862900, NCT01303705).
3.2. CD137
CD137 (4-1BB) is an inducible T-cell surface molecule of the tumor necrosis factor (TNF) receptor superfamily. Binding to its ligand CD137L co-stimulates CD4 and CD8 cells [82]. CD137 is also expressed on Treg, activated NK cells. Urelumab (BMS 663513) a CD137 agonist [83] has been tested in a phase I/II study with promising activity although marked hepatic toxicities has been reported [83,84]. A study of urelumab in NSCLC has been terminated (NCT00461110).
3.3. GITR
Glucocorticoid-induced TNF receptor (GITR) is a member of the TNF receptor superfamily. GITR acts as a co-stimulatory molecule to both CD4+ and CD8+ naïve T cells, leading to T cell proliferation and effector function [85]. GITR is found on Treg, effector T cells, B cells, NK cells, and activated dendritic cells [86]. TRX518, a monoclonal antibody GITR agonist, is being investigated in a phase I setting (NCT01239134).
3.4. CD40, CD28, CD27 and ICOS
CD40 is a member of the tumor necrosis factor superfamily and is involved in cellular differentiation, survival and apoptosis [87]. Preclinical studies have shown a potential use of anti-CD40 antibodies may suppress tumor growth and metastases [88]. CD27 is another stimulatory receptor of T cells and an antibody against it is currently being tested in a phase I study (NCT01460134). Inducible T-cell costimulator (ICOS) is a T-cell co-stimulator related to CD28 [89]. TGN1412, an antibody against CD28 was found to be extremely toxic and has hence no further trials have been conducted with this class of drugs [90].
4. Combination therapy
The concept of using multiple drugs to target the various pathways of tumorigenesis has been a common approach utilized in cancer therapy. This includes the use of combination cytotoxic chemotherapeutic agents with different mechanisms of action and toxicity profiles to maximize cell kill. Indeed, the standard of care for first line chemotherapy regimen for NSCLC is a platinum based doublet [91]. With the emergence of molecular-targeted therapy in NSCLC, it has become increasing apparent that combinations require careful investigation in their consideration. While some combinations of targeted therapy and chemotherapy have been found to be of benefit, other combinations may be detrimental [92]. While, each agent has proven to be efficacious individually, the combination of these agents has not shown to improve overall survival. Similarly, especially with its distinctive toxicity profile, the combination of immunotherapeutic agents with one another and with chemotherapy regimens to affect the multiple aspects of the immunomodulation of the neoplastic process requires further study.
CTLA4 inhibitors in combination with chemotherapy have already been discussed previously. Currently there is a phase III study looking at the role of ipilimumab with paclitaxel and carboplatin in squamous NSCLC (NCT01285609). Nivolumab and Ipilimumab in combination with KIR inhibitors (NCT01714739, NCT01750580), and nivolumab in combination with various chemotherapeutic and targeted therapies (NCT01454102) has also been described above (Table 3).
Of interest is the combination of immunotherapeutic agents with each other, and currently several are being tested. CTLA4 blockade after vaccination with irradiated, autologous tumor cells engineered to secrete granulocyte-macrophage colony stimulating factor (GM-CSF) showed antitumor immunity without major toxicities in patients with advanced melanoma [93]. Multiple immune checkpoint blockades with combination PD1 and CTLA4 Ab can allow for increased T cell responsiveness and decreased T cell anergy, in preclinical models [94].
The timing of immunotherapy in the schema of anti-neoplastic therapy requires further study. In the phase II study of ipilimumab and chemotherapy described earlier, the arm which introduced ipilimumab in a delayed manner compared to the upfront introduction of ipilimumab appeared to have a greater benefit, and the biology of this is not clearly understood yet.
5. Predictive biomarkers
The need for predictive biomarkers is important as immune modulators can result in durable response but only in a very select group of patients. Several biomarkers for CTLA4 have been described that may translate into clinical application [95] A rise in absolute lymphocyte counts has been shown to be associated with clinical benefit [96]. Studies have shown a positive correlation between ipilimumab treatment and frequency of CD4+ cells expressing high levels of ICOS [97]. HLA-DR has been associated with increased expression on CD4 cells after treatment with ipilimumab [98] Similar results have also been shown with CD45RO [99]. Tumor infiltrating lymphocytes have been found to have correlate with clinical benefit from treatment with ipilimumab [100] Forkhead box P3 (FOXP3)-positive and indoleamine 2,3-dioxygenase (IDO)-positive patients benefited from CTLA4 blockage as well [100].
It is known that PD-L1 expression varies with the tumor microenvironment and may not be a static expression at a single time point. PD-L1 expression has been associated with the presence of IFN-γ in the tumor microenvironment [101]. This may be the explanation for PD-L1 expression not being a good predictive marker for nivolumab as described earlier [41]. Predictive biomarker research in this area is at an early stage of development and multiple studies are ongoing examining issues such as the following: Is a predictive biomarker/companion diagnostic necessary? Is tissue micro-localization of biomarker expression important (TILS versus tumor)? What is the optimal detection method? Does biomarker expression alter over time (time at diagnosis versus time at relapse) and space (primary versus metastasis)? What is the impact of chemotherapy, molecular targeted therapy and radiotherapy on biomarker expression?
6. Evaluation of tumor response to immunotherapeutic agents
One accepted standardized method to evaluate tumor response to cancer treatment is the use of RECIST criteria [102]. However, according to RECIST, an increase in the size of a tumor of 30% or more, even early in treatment, is considered as disease progression. However treatment responses with immunotherapy may occur even after progression of disease as defined by RECIST criteria. Patients appear to have clinical benefit even though the tumor appears to progress radiologically and sustained SD may represent sustained response. Based on this, an immune related Response Criteria has been created [103]. Immune related progression free survival (irPFS) accounts for the apparent increase in tumor size followed by sustained tumor response which has been documented with these agents in the past [104]. This phenomenon of “pseudo-progression” may be due to peritumoral lymphocyte infiltration or delayed immune activity.
7. Conclusion
An improvement in the understanding of the immune system in tumor immunosurveillance has resulted in the development of a new generation of immunotherapeutic agents. In particular, it is now recognized tumors can evade immune destruction via the dysregulation of co-inhibitory or checkpoint signals. Results from early phase studies of immune checkpoint modulators such as CTLA-4, PD-1 and PD-L1 inhibitors in a range of solid tumors including NSCLC are highly promising and may provide new therapeutic options in the treatment of NSCLC. Future challenges include incorporating immunotherapeutic agents either together or in combination with cytotoxic chemotherapy, molecular targeted therapy and radiotherapy, its administration in early stage disease and the identification of predictive biomarkers.
Acknowledgments
RAS is supported by the National Research Foundation Singapore and the Singapore Ministry of Education under its Research Centres of Excellence initiative.
Footnotes
Conflict of interest statement
Byoung-Chul Cho has received honoraria from AstraZeneca, Pfizer, Boehringer-Ingelheim, GSK, and Novartis; research grants from AstraZeneca, Novartis, Bayer, Boehringer-Ingelheim. Julie R. Brahmer is an uncompensated advisory board member for BMS, is an advisory board member for Merck, and her institution has received a BMS research grant. Ross Soo has received honoraria from Astra-Zeneca, Roche, Norvatis and Boehringer-Ingelheim. Raghav Sundar and Richie Soong have no conflict of interest.
References
- 1.Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
- 2.Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57. doi: 10.1056/NEJMoa0810699. [DOI] [PubMed] [Google Scholar]
- 3.Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12:175–80. doi: 10.1016/S1470-2045(10)70087-5. [DOI] [PubMed] [Google Scholar]
- 4.Schiller JH, Morgan-Ihrig C, Levitt ML. Concomitant administration of interleukin-2 plus tumor necrosis factor in advanced non-small cell lung cancer. Am J Clin Oncol. 1995;18:47–51. doi: 10.1097/00000421-199502000-00010. [DOI] [PubMed] [Google Scholar]
- 5.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 6.Einhorn LH, Bond WH, Hornback N, Joe BT. Long-term results in combined-modality treatment of small cell carcinoma of the lung. Semin Oncol. 1978;5:309–13. [PubMed] [Google Scholar]
- 7.Jansen RL, Slingerland R, Goey SH, Franks CR, Bolhuis RL, Stoter G. Interleukin-2 and interferon-alpha in the treatment of patients with advanced non-small-cell lung cancer. J Immunother (1991) 1992;12:70–3. doi: 10.1097/00002371-199207000-00009. [DOI] [PubMed] [Google Scholar]
- 8.Butts C, Socinski MA, Mitchell PL, Thatcher N, Havel L, Krzakowski M, et al. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15:59–68. doi: 10.1016/S1470-2045(13)70510-2. [DOI] [PubMed] [Google Scholar]
- 9.Giaccone G, Bazhenova L, Nemunaitis J, Juhasz E, Ramlau R, van den Heuvel M, et al. A phase III study of belagenpumatucel-L therapeutic tumor cell vaccine for non-small cell lung cancer. European Cancer Congress. 2013 doi: 10.1016/j.ejca.2015.07.035. Abstract LBA 2. [DOI] [PubMed] [Google Scholar]
- 10.Media release. Investigational MAGE-A3 antigen-specific cancer immunotherapeutic does not meet first co-primary endpoints in MAGRIT, a phase III non-small cell lung cancer clinical trial. 2014 http://www.gsk.com/media/press-releases/2014/investigational-MAGE-A3-antigen-specificcancer-immunotherapeutic-does-not-meet-first-co-primary-endpoints-in-MAGRIT.html (accessed 30.03.14)
- 11.Mellstedt H, Vansteenkiste J, Thatcher N. Vaccines for the treatment of non-small cell lung cancer: investigational approaches and clinical experience. Lung Cancer. 2011;73:11–7. doi: 10.1016/j.lungcan.2011.02.023. [DOI] [PubMed] [Google Scholar]
- 12.O’Day SJ, Hamid O, Urba WJ. Targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4): a novel strategy for the treatment of melanoma and other malignancies. Cancer. 2007;110:2614–27. doi: 10.1002/cncr.23086. [DOI] [PubMed] [Google Scholar]
- 13.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nishimura H, Honjo T. PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol. 2001;22:265–8. doi: 10.1016/s1471-4906(01)01888-9. [DOI] [PubMed] [Google Scholar]
- 15.Roth CG, Garner K, Eyck ST, Boyiadzis M, Kane LP, Craig FE. TIM3 expression by leukemic and non-leukemic myeloblasts. Cytometry B Clin Cytom. 2013;84:167–72. doi: 10.1002/cyto.b.21080. [DOI] [PubMed] [Google Scholar]
- 16.Wada J, Kanwar YS. Identification and characterization of galectin-9, a novel beta-galactoside-binding mammalian lectin. J Biol Chem. 1997;272:6078–86. doi: 10.1074/jbc.272.9.6078. [DOI] [PubMed] [Google Scholar]
- 17.Sega EI, Leveson-Gower DB, Florek M, Schneidawind D, Luong RH, Negrin RS. Role of lymphocyte activation gene-3 (lag-3) in conventional and regulatory T cell function in allogeneic transplantation. PLOS ONE. 2014;9:e86551. doi: 10.1371/journal.pone.0086551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Watanabe N, Gavrieli M, Sedy JR, Yang J, Fallarino F, Loftin SK, et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat Immunol. 2003;4:670–9. doi: 10.1038/ni944. [DOI] [PubMed] [Google Scholar]
- 19.Pasero C, Olive D. Interfering with coinhibitory molecules: BTLA/HVEM as new targets to enhance anti-tumor immunity. Immunol Lett. 2013;151:71–5. doi: 10.1016/j.imlet.2013.01.008. [DOI] [PubMed] [Google Scholar]
- 20.Lee CF, Lai HL, Lee YC, Chien CL, Chern Y. The A2A adenosine receptor is a dual coding gene: a novel mechanism of gene usage and signal transduction. J Biol Chem. 2014;289:1257–70. doi: 10.1074/jbc.M113.509059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sica GL, Choi IH, Zhu G, Tamada K, Wang SD, Tamura H, et al. B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity. 2003;18:849–61. doi: 10.1016/s1074-7613(03)00152-3. [DOI] [PubMed] [Google Scholar]
- 22.Kohrt HE, Thielens A, Marabelle A, Sagiv-Barfi I, Sola C, Chanuc F, et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood. 2014;123:678–86. doi: 10.1182/blood-2013-08-519199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Farres MN, Sabry MK, Ahmed EE, Elkady HM, Mohamed NA. OX40 ligand: a potential costimulatory molecule in atopic asthma. J Asthma. 2014 doi: 10.3109/02770903.2014.897729. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 24.Shao Z, Schwarz H. CD137 ligand, a member of the tumor necrosis factor family, regulates immune responses via reverse signal transduction. J Leukoc Biol. 2011;89:21–9. doi: 10.1189/jlb.0510315. [DOI] [PubMed] [Google Scholar]
- 25.Chatzigeorgiou A, Lyberi M, Chatzilymperis G, Nezos A, Kamper E. CD40/CD40L signaling and its implication in health and disease. Biofactors. 2009;35:474–83. doi: 10.1002/biof.62. [DOI] [PubMed] [Google Scholar]
- 26.Viac J, Schmitt D, Claudy A. CD40 expression in epidermal tumors. Anticancer Res. 1997;17:569–72. [PubMed] [Google Scholar]
- 27.Prasad KV, Ao Z, Yoon Y, Wu MX, Rizk M, Jacquot S, et al. CD27, a member of the tumor necrosis factor receptor family, induces apoptosis and binds to Siva, a proapoptotic protein. Proc Natl Acad Sci U S A. 1997;94:6346–51. doi: 10.1073/pnas.94.12.6346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Nocentini G, Ronchetti S, Cuzzocrea S, Riccardi C. GITR/GITRL: more than an effector T cell co-stimulatory system. Eur J Immunol. 2007;37:1165–9. doi: 10.1002/eji.200636933. [DOI] [PubMed] [Google Scholar]
- 29.Yoshinaga SK, Whoriskey JS, Khare SD, Sarmiento U, Guo J, Horan T, et al. T-cell co-stimulation through B7RP-1 and ICOS. Nature. 1999;402:827–32. doi: 10.1038/45582. [DOI] [PubMed] [Google Scholar]
- 30.Woerly G, Roger N, Loiseau S, Dombrowicz D, Capron A, Capron M. Expression of CD28 and CD86 by human eosinophils and role in the secretion of type 1 cytokines (interleukin 2 and interferon gamma): inhibition by immunoglobulin a complexes. J Exp Med. 1999;190:487–95. doi: 10.1084/jem.190.4.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol. 2001;19:565–94. doi: 10.1146/annurev.immunol.19.1.565. [DOI] [PubMed] [Google Scholar]
- 32.Thompson CB, Allison JP. The emerging role of CTLA-4 as an immune attenuator. Immunity. 1997;7:445–50. doi: 10.1016/s1074-7613(00)80366-0. [DOI] [PubMed] [Google Scholar]
- 33.Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, et al. Lymphoproliferative disorders with early lethality in mice deficient in CTLA-4. Science. 1995;270:985–8. doi: 10.1126/science.270.5238.985. [DOI] [PubMed] [Google Scholar]
- 34.Salvi S, Fontana V, Boccardo S, Merlo DF, Margallo E, Laurent S, et al. Evaluation of CTLA-4 expression and relevance as a novel prognostic factor in patients with non-small cell lung cancer. Cancer Immunol Immunother. 2012;61:1463–72. doi: 10.1007/s00262-012-1211-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zheng H, Li Y, Wang X, Zhang X, Wang X. Expression and significance of gp96 and immune-related gene CTLA-4, CD8 in lung cancer tissues. Zhongguo Fei Ai Za Zhi. 2010;13:790–4. doi: 10.3779/j.issn.1009-3419.2010.08.08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. 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;30:2046–54. doi: 10.1200/JCO.2011.38.4032. [DOI] [PubMed] [Google Scholar]
- 37.Zatloukal P, Heo DS, Park K, Kang J, Butts C, Bradford D, et al. Randomized phase II clinical trial comparing tremelimumab (CP-675,206) with best supportive care (BSC) following first-line platinum-based therapy in patients (pts) with advanced non-small cell lung cancer (NSCLC) J Clin Oncol. 2009;27 (suppl; abstr 15S, 8071) [Google Scholar]
- 38.Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Brahmer J, Horn L, Antonia S, Spigel D, Gandhi L, Sequist L, et al. Nivolumab (anti-PD-1; BMS-936558; ONO-4538) in patients with non-small cell lung cancer (NSCLC): overall survival and long-term safety in a phase 1 trial. IASLC 15th World Conference on Lung Cancer. 2013 Abstract MO18.03. [Google Scholar]
- 41.Antonia S, Grosso J, Horak C, Harbison C, Kurland J, Inzunza D, et al. Association of tumor PD-L1 expression and immune biomarkers with clinical activity in patients with non-small cell lung cancer treated with nivolumab. IASLC 15th World Conference on Lung Cancer. 2013 Abstract P2.11–035. [Google Scholar]
- 42.Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Garon E, Balmanoukian A, Hamid O, Hui R, Gandhi L, Leighi N, et al. Preliminary clinical safety and activity of MK-3475 mono-therapy for the treatment of previously treated patients with non-small cell lung cancer. IASLC 15th World Conference on Lung Cancer. 2013 Abstract MO18.02. [Google Scholar]
- 44.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
- 45.Yang CY, Lin MW, Chang YL, Wu CT, Yang PC. Programmed cell death-ligand 1 expression in surgically resected stage I pulmonary adenocarcinoma and its correlation with driver mutations and clinical outcomes. Eur J Cancer. 2014;50:1361–9. doi: 10.1016/j.ejca.2014.01.018. [DOI] [PubMed] [Google Scholar]
- 46.Velcheti V, Schalper KA, Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, et al. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Invest. 2014;94:107–16. doi: 10.1038/labinvest.2013.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chen YY, Wang LB, Zhu HL, Li XY, Zhu YP, Yin YL, et al. Relationship between programmed death-ligand 1 and clinicopathological characteristics in non-small cell lung cancer patients. Chin Med Sci J. 2013;28:147–51. doi: 10.1016/s1001-9294(13)60040-1. [DOI] [PubMed] [Google Scholar]
- 48.Chen YB, Mu CY, Huang JA. Clinical significance of programmed death-1 ligand-1 expression in patients with non-small cell lung cancer: a 5-year-follow-up study. Tumori. 2012;98:751–5. doi: 10.1177/030089161209800612. [DOI] [PubMed] [Google Scholar]
- 49.Boland JM, Kwon ED, Harrington SM, Wampfler JA, Tang H, Yang P, et al. Tumor B7-H1 and B7-H3 expression in squamous cell carcinoma of the lung. Clin Lung Cancer. 2013;14:157–63. doi: 10.1016/j.cllc.2012.05.006. [DOI] [PubMed] [Google Scholar]
- 50.Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol. 2011;28:682–8. doi: 10.1007/s12032-010-9515-2. [DOI] [PubMed] [Google Scholar]
- 51.Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res. 2004;10:5094–100. doi: 10.1158/1078-0432.CCR-04-0428. [DOI] [PubMed] [Google Scholar]
- 52.Velcheti V, Rimm DL, Schalper KA. Sarcomatoid lung carcinomas show high levels of programmed death ligand-1 (PD-L1) J Thorac Oncol. 2013;8:803–5. doi: 10.1097/JTO.0b013e318292be18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Soria J, Cruz C, Bahleda R, Delord J, Horn L, Herbst R, et al. Clinical activity, safety and biomarkers of PD-L1 blockade in non-small cell lung cancer (NSCLC): additional analyses from a clinical study of the engineered antibody MPDL3280A (anti-PDL1) European Cancer Congress. 2013 Abstract 3408. [Google Scholar]
- 55.Khleif S, Lutzky J, Segal N, Antonia S, Blake-Haskins A, Stewart R, et al. MEDI4736, an anti-PD-L1 antibody with modified Fc domain: preclinical evaluation and early clinical results from a phase 1 study in patients with advanced solid tumors. European Cancer Congress. 2013 Abstract 802. [Google Scholar]
- 56.Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415:536–41. doi: 10.1038/415536a. [DOI] [PubMed] [Google Scholar]
- 57.Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6:1245–52. doi: 10.1038/ni1271. [DOI] [PubMed] [Google Scholar]
- 58.Sabatos CA, Chakravarti S, Cha E, Schubart A, Sanchez-Fueyo A, Zheng XX, et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol. 2003;4:1102–10. doi: 10.1038/ni988. [DOI] [PubMed] [Google Scholar]
- 59.Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207:2187–94. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207:2175–86. doi: 10.1084/jem.20100637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gao X, Zhu Y, Li G, Huang H, Zhang G, Wang F, et al. TIM-3 expression charac-terizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS ONE. 2012;7:e30676. doi: 10.1371/journal.pone.0030676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhuang X, Zhang X, Xia X, Zhang C, Liang X, Gao L, et al. Ectopic expression of TIM-3 in lung cancers: a potential independent prognostic factor for patients with NSCLC. Am J Clin Pathol. 2012;137:978–85. doi: 10.1309/AJCP9Q6OVLVSHTMY. [DOI] [PubMed] [Google Scholar]
- 63.Zhou Q, Munger ME, Veenstra RG, Weigel BJ, Hirashima M, Munn DH, et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood. 2011;117:4501–10. doi: 10.1182/blood-2010-10-310425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ngiow SF, von Scheidt B, Akiba H, Yagita H, Teng MW, Smyth MJ. Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors. Cancer Res. 2011;71:3540–51. doi: 10.1158/0008-5472.CAN-11-0096. [DOI] [PubMed] [Google Scholar]
- 65.Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21:503–13. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
- 66.Gandhi MK, Lambley E, Duraiswamy J, Dua U, Smith C, Elliott S, et al. Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. Blood. 2006;108:2280–9. doi: 10.1182/blood-2006-04-015164. [DOI] [PubMed] [Google Scholar]
- 67.Freire J, Ajona D, de Biurrun G, Agorreta J, Segura V, Guruceaga E, et al. Silica-induced chronic inflammation promotes lung carcinogenesis in the context of an immunosuppressive microenvironment. Neoplasia. 2013;15:913–24. doi: 10.1593/neo.13310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
- 69.Romagne F, Andre P, Spee P, Zahn S, Anfossi N, Gauthier L, et al. Preclinical characterization of 1-7F9, a novel human anti-KIR receptor therapeutic antibody that augments natural killer-mediated killing of tumor cells. Blood. 2009;114:2667–77. doi: 10.1182/blood-2009-02-206532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Al Omar S, Middleton D, Marshall E, Porter D, Xinarianos G, Raji O, et al. Associations between genes for killer immunoglobulin-like receptors and their ligands in patients with solid tumors. Hum Immunol. 2010;71:976–81. doi: 10.1016/j.humimm.2010.06.019. [DOI] [PubMed] [Google Scholar]
- 71.Gertner-Dardenne J, Fauriat C, Orlanducci F, Thibult ML, Pastor S, Fitzgibbon J, et al. The co-receptor BTLA negatively regulates human Vgamma9Vdelta2 T-cell proliferation: a potential way of immune escape for lymphoma cells. Blood. 2013;122:922–31. doi: 10.1182/blood-2012-11-464685. [DOI] [PubMed] [Google Scholar]
- 72.Paterson DJ, Jefferies WA, Green JR, Brandon MR, Corthesy P, Puklavec M, et al. Antigens of activated rat T lymphocytes including a molecule of 50,000 Mr detected only on CD4 positive T blasts. Mol Immunol. 1987;24:1281–90. doi: 10.1016/0161-5890(87)90122-2. [DOI] [PubMed] [Google Scholar]
- 73.Gramaglia I, Weinberg AD, Lemon M, Croft M. Ox-40 ligand: a potent cos-timulatory molecule for sustaining primary CD4 T cell responses. J Immunol. 1998;161:6510–7. [PubMed] [Google Scholar]
- 74.Sugamura K, Ishii N, Weinberg AD. Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40. Nat Rev Immunol. 2004;4:420–31. doi: 10.1038/nri1371. [DOI] [PubMed] [Google Scholar]
- 75.Weinberg AD, Rivera MM, Prell R, Morris A, Ramstad T, Vetto JT, et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol. 2000;164:2160–9. doi: 10.4049/jimmunol.164.4.2160. [DOI] [PubMed] [Google Scholar]
- 76.Morris A, Vetto JT, Ramstad T, Funatake CJ, Choolun E, Entwisle C, et al. Induction of anti-mammary cancer immunity by engaging the OX-40 receptor in vivo. Breast Cancer Res Treat. 2001;67:71–80. doi: 10.1023/a:1010649303056. [DOI] [PubMed] [Google Scholar]
- 77.Ali SA, Ahmad M, Lynam J, McLean CS, Entwisle C, Loudon P, et al. Antitumour therapeutic efficacy of OX40L in murine tumour model. Vaccine. 2004;22:3585–94. doi: 10.1016/j.vaccine.2004.03.041. [DOI] [PubMed] [Google Scholar]
- 78.Sadun RE, Hsu WE, Zhang N, Nien YC, Bergfeld SA, Sabzevari H, et al. Fc-mOX40L fusion protein produces complete remission and enhanced survival in 2 murine tumor models. J Immunother. 2008;31:235–45. doi: 10.1097/CJI.0b013e31816a88e0. [DOI] [PubMed] [Google Scholar]
- 79.Redmond WL, Gough MJ, Weinberg AD. Ligation of the OX40 co-stimulatory receptor reverses self-Ag and tumor-induced CD8 T-cell anergy in vivo. Eur J Immunol. 2009;39:2184–94. doi: 10.1002/eji.200939348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, et al. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013;73:7189–98. doi: 10.1158/0008-5472.CAN-12-4174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Morris NP, Peters C, Montler R, Hu HM, Curti BD, Urba WJ, et al. Development and characterization of recombinant human Fc:OX40L fusion protein linked via a coiled-coil trimerization domain. Mol Immunol. 2007;44:3112–21. doi: 10.1016/j.molimm.2007.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, et al. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest. 2002;109:651–9. doi: 10.1172/JCI14184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ascierto PA, Marincola FM, Ribas A. Anti-CTLA4 monoclonal antibodies: the past and the future in clinical application. J Transl Med. 2011;9:196. doi: 10.1186/1479-5876-9-196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sznol M, Hodi FS, Margolin K, McDermott DF, Ernstoff MS, Kirkwood JM, et al. Phase I study of BMS-663513, a fully human anti-CD137 agonist monoclonal antibody, in patients (pts) with advanced cancer (CA) J Clin Oncol. 2008;26 (suppl; abstr 3007) [Google Scholar]
- 85.Ronchetti S, Nocentini G, Riccardi C, Pandolfi PP. Role of GITR in activation response of T lymphocytes. Blood. 2002;100:350–2. doi: 10.1182/blood-2001-12-0276. [DOI] [PubMed] [Google Scholar]
- 86.Arnett SO, Teillaud JL, Wurch T, Reichert JM, Dunlop C, Huber M. IBC’s 21stannual antibody engineering and 8th annual antibody therapeutics inter-national conferences and 2010 annual meeting of the antibody society. December 5–9, 2010, San Diego, CA, USA. MAbs. 2011;3:133–52. doi: 10.4161/mabs.3.2.14939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sabel MS, Yamada M, Kawaguchi Y, Chen FA, Takita H, Bankert RB. CD40 expression on human lung cancer correlates with metastatic spread. Cancer Immunol Immunother. 2000;49:101–8. doi: 10.1007/s002620050608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.von Scheidt B, Leung PS, Yong MC, Zhang Y, Towne JE, Smyth MJ, et al. Combined anti-CD40 and anti-IL-23 monoclonal antibody therapy effectively suppresses tumor growth and metastases. Cancer Res. 2014;74:2412–21. doi: 10.1158/0008-5472.CAN-13-1646. [DOI] [PubMed] [Google Scholar]
- 89.Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature. 1999;397:263–6. doi: 10.1038/16717. [DOI] [PubMed] [Google Scholar]
- 90.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018–28. doi: 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
- 91.Delbaldo C, Michiels S, Syz N, Soria JC, Le Chevalier T, Pignon JP. Benefits of adding a drug to a single-agent or a 2-agent chemotherapy regimen in advanced non-small-cell lung cancer: a meta-analysis. JAMA. 2004;292:470–84. doi: 10.1001/jama.292.4.470. [DOI] [PubMed] [Google Scholar]
- 92.Herbst RS, Prager D, Hermann R, Fehrenbacher L, Johnson BE, Sandler A, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol. 2005;23:5892–9. doi: 10.1200/JCO.2005.02.840. [DOI] [PubMed] [Google Scholar]
- 93.Hodi FS, Butler M, Oble DA, Seiden MV, Haluska FG, Kruse A, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A. 2008;105:3005–10. doi: 10.1073/pnas.0712237105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA. 2010;107:4275–80. doi: 10.1073/pnas.0915174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Callahan MK, Wolchok JD, Allison JP. Anti-CTLA-4 antibody therapy: immune monitoring during clinical development of a novel immunotherapy. Semin Oncol. 2010;37:473–84. doi: 10.1053/j.seminoncol.2010.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Berman DM, Wolchok J, Weber J, Hamid O, O’Day S, Chasalow SD. Association of peripheral blood absolute lymphocyte count (ALC) and clinical activity in patients (pts) with advanced melanoma treated with ipilimumab. J Clin Oncol. 2009;27 (suppl; abstr 3020) [Google Scholar]
- 97.Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A. 2008;105:14987–92. doi: 10.1073/pnas.0806075105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100:8372–7. doi: 10.1073/pnas.1533209100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Maker AV, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, et al. Intra-patient dose escalation of anti-CTLA-4 antibody in patients with metastatic melanoma. J Immunother. 2006;29:455–63. doi: 10.1097/01.cji.0000208259.73167.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hamid O, Schmidt H, Nissan A, Ridolfi L, Aamdal S, Hansson J, et al. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med. 2011;9:204. doi: 10.1186/1479-5876-9-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4:127ra37. doi: 10.1126/scitranslmed.3003689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–47. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
- 103.Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–20. doi: 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
- 104.Oxnard GR, Morris MJ, Hodi FS, Baker LH, Kris MG, Venook AP, et al. When progressive disease does not mean treatment failure: reconsidering the criteria for progression. J Natl Cancer Inst. 2012;104:1534–41. doi: 10.1093/jnci/djs353. [DOI] [PMC free article] [PubMed] [Google Scholar]
