Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2019

Open Access 01.12.2019 | Review

Potential immune escape mechanisms underlying the distinct clinical outcome of immune checkpoint blockades in small cell lung cancer

verfasst von: Yaru Tian, Xiaoyang Zhai, Anqin Han, Hui Zhu, Jinming Yu

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2019

Abstract

Small cell lung cancer (SCLC) is one of the deadliest cancer types in the world. Despite the high response rate to frontline platinum-containing doublets, relapse is inevitable for the majority of patients and the prognosis is poor. Topotecan, which has limited efficacy, has remained the standard second-line therapy for approximately three decades. Although SCLC has a high mutation burden, the clinical efficacy of immune checkpoint blockades (ICBs) in SCLC is far less pronounced than that in non-small cell lung cancer (NSCLC). Only atezolizumab in combination with chemotherapy improved overall survival over chemotherapy alone in the phase III CheckMate 133 trial and has recently received FDA approval as first-line therapy. Most studies concerning ICBs in SCLC are limited to early-phase studies and found that ICBs were not superior to traditional chemotherapy. Why is there such a large difference between SCLC and NSCLC? In this review, comparative analyses of previous studies indicate that SCLC is even more immunodeficient than NSCLC and the potential immune escape mechanisms in SCLC may involve the low expression of PD-L1 and the downregulation of major histocompability complex (MHC) molecules and regulatory chemokines. In consideration of these immune dysfunctions, we speculate that chemotherapy and radiotherapy prior to immunotherapy, the combination of ICBs with antiangiogenic treatment, and selecting tumor mutation burden in combination with PD-L1 expression as biomarkers could be promising strategies to improve the clinical efficacy of immunotherapy for SCLC.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AACR
American Association for Cancer Research
AE
Adverse event
CFDA
Chinese Food and Drug Administration
CTLA4
Cytotoxic T-lymphocyte protein-4
ES-SCLC
Extensive-stage SCLC
FDA
US Food and Drug Administration
G3-4
Grade 3-4
GCP-2
Granulocyte chemotactic protein
ICAM
Intercellular adhesion molecule-1
ICB
Immune checkpoint blockade
INF-γ
Interferon-γ
ir
Immune-related
LS-SCLC
Limited-stage SCLC
MHC
Major histocompability complex
NSCLC
Non-small cell lung cancer
ORR
Overall response rate
OS
Overall survival
PD-1
Programmed cell death protein-1
PD-L1
Programmed cell death ligand-1
PFS
Progression-free survival
SCF
Stem cell factor
SCLC
Small cell lung cancer
TIL
Tumor infiltration lymphocyte
TIM
Tumor infiltration macrophage
TMB
Tumor mutation burden
TME
Tumor mutation burden

Background

Worldwide, lung cancer is the leading cause of cancer incidence and mortality, with 2.1 million new cases and 1.8 million deaths estimated in 2018, representing 18.4% of the total cancer-related deaths [1]. Small cell lung cancer (SCLC) accounts for approximately 14% of all lung cancers [2, 3] and is highly aggressive and lethal, characterized by a rapid growth rate and early development of widespread metastasis. Most patients with SCLC have a history of cigarette smoking [46] and harbor a high prevalence of somatic mutations [710]. SCLC is classified as limited-stage SCLC (LS-SCLC) and extensive-stage SCLC (ES-SCLC). ES-SCLC, generally with distant metastasis at the time of diagnosis, accounts for approximately two thirds of all SCLC. Platinum-based chemotherapy remains the first-line standard of care for SCLC and the response rate could reach more than 70%. Although SCLC is highly responsive to initial therapy, most patients with SCLC will inevitably relapse. Consequently, the prognosis of SCLC is rather poor, with an estimated 2-year overall survival (OS) of less than 5% [1115]. Currently, topotecan is the only drug approved by the US Food and Drug Administration (FDA) as a second-line therapy. However, the efficacy of topotecan is limited, with a response rate of approximately 25% for platinum-sensitive patients and less than 10% for platinum-resistant or refractory patients [1618]. Other recommended second-line options included irinotecan, vinorelbine, gemcitabine, and temozolomide [11]. Clinical benefit is even dismal for treatment beyond the second line. Contrasting sharply with non-small cell lung cancer (NSCLC), SCLC is a hard-to-treat cancer with lack of progress for nearly 30 years.
Immunotherapy has revolutionized the standard of care across multiple cancer types. As tumors develop, they can escape immune surveillance by various mechanisms in steps in the cancer-immunity cycle [19, 20]. Immune checkpoints, limiting antitumor responses and contributing to immune escape, have been readily confirmed as negative regulators in recent years. Two such checkpoints, cytotoxic T-lymphocyte protein 4 (CTLA4) and programmed cell death protein-1 (PD-1), are the best [21]. The co-inhibitory receptor CTLA-4, which is expressed on T cells, can outcompete CD28 to bind to CD80 and CD86 on APCs preventing both the activation and proliferation of T cells in the lymphnodes [22, 23]. Moreover, the highly constitutively expressed CTLA-4 on Treg cells also mediates the suppressive role of Treg cells. Anti-CTLA4 blockades significantly improve the overall survival of patients with advanced melanoma [24, 25]. Differing from CTLA4 in its mechanism of action, the PD-1/PD-L1 pathway mainly mediates T cell dysfunction in the tumor microenvironment (TME). PD-1 is induced on activated T cells through TCR and co-stimulatory signals. These effector T cells are capable of recognizing tumor neoantigens and then producing interferon-γ (INF-γ), thereby inducing the expression of PD-L1 on tumor cells and stromal cells. Ultimately, the interaction between PD-L1 and PD-1 disrupts the antitumor activity of effector T cells. This local immune escape mechanism has been termed “adaptive immune resistance” [2629]. Sanmamed and Chen [30] have confirmed anti-PD/PD-L1 therapy as a clearest approach to the normalization of cancer immunotherapy; it can selectively repair the tumor-induced immune defect and restore immune activity in the TME without general immune activation [30]. Patients with a variety of cancer types have exhibited promising clinical benefit/risk ratio from anti-PD-1/PD-L1 therapies, resulting in FDA approval for the corresponding indications [21, 26, 30, 31].
The progress made in uncovering the biology of SCLC and its microenvironment has offered new therapeutic strategies for SCLC [32]. The high mutation burden, which produces a large number of potential tumor-specific antigens, raised hope regarding immunotherapy in SCLC [33]. In this review, we mainly illustrate the rationale behind immunotherapy for SCLC, the differences in clinical outcomes of ICBs and the underlying mechanisms in NSCLC and SCLC. Finally, we propose some strategies to improve the clinical outcome of immunotherapy for SCLC.

Rationale of immunotherapy therapy for SCLC

Nearly 98% of SCLC patients have a smoking history [46]. Tobacco can exert carcinogenic effects through more than 60 chemicals that are capable of binding and mutating DNA [10]. Consequently, SCLC has a high tumor mutation burden (TMB) of 8.62 nonsynonymous or missense mutations per million base pairs among various solid tumors [710]. These mutations can therefore generate neoantigens (new protein or peptide sequences) that will be presented by MHC molecules and recognized by T cells [20, 34, 35]. Immunogenic neoantigens that elicit tumor-specific CD8+ T cell activities can thereby enhance the clinical benefits of immune checkpoint blockades (ICBs) [36, 37]. The TMB has been confirmed as an effective biomarker to predict the clinical outcome of ICBs in many tumors [35, 38]. Moreover, there is a similar level and distribution of TMBs between NSCLC and SCLC, and similar to in NSCLC, the TMB has been associated with the clinical outcome of ICBs in SCLC [33, 39].
Moreover, retrospective findings indicated that host immune status was strongly associated with the prognosis of SCLC. The inflammatory status of the host immune system was suggestive of a beneficial clinical outcome. Immunologic indicators associated with better prognosis included tumor-infiltrating macrophages (TIMs) and lymphocytes (TILs) [4042], effective CD4+ T cells including Th17 cells [43], a high effector T cell (Teff) to regulatory T cell (Treg) ratio [43], a high neutrophil to lymphocyte ratio (NLR) and a high platelet to lymphocyte ratio (PLR) [44]. In addition, suppressive immune features indicating a poor prognosis of SCLC include the frequency of CD14+HLA-DR-/low myeloid-derived suppressor cells (MDSCs) [45], the C-reactive protein/albumin (CRP/Alb) ratio[46] and a higher Treg cell ratio in tumor infiltrates [47]. These clinical evidence further illustrated the significance of the immune response and the possibility of immunotherapy for SCLC.
In conclusion, the high TMB and the status of the host immune system may hold new promise for immunotherapy for SCLC. Among multiple immunotherapy strategies, ICBs have achieved great success in treating SCLC.

Clinical outcomes of ICBs in NSCLC and SCLC

Immune checkpoint blockades, including anti-CTLA4 antibody (ipilimumab), anti-PD-1 antibodies (pembrolizumab and nivolumab), and anti-PD-L1 antibodies (atezolizumab and durvalumab), have showed remarkable and durable responses across multiple cancer types and received FDA approval [21, 26, 30, 31]. Table 1 summarizes the major clinical trials involving ICBs in NSCLC and SCLC.
Table 1
Comparisons of clinical outcomes of ICBs between NSCLC and SCLC
NSCLC
SCLC
Study
Design
ORR, PFS, and OS
TRAE
Study
Design
ORR, PFS, and OS
TRAE
First-line
NCT 00527735 [48]
II R 1: 1: 1
N = 204
1. Placebo+Pacl+Carbo
2. Concurrent Ipi+Pacl+Carbo
3. Phased Ipi+Pacl+Carbo
irBORR, 18%, 21%, 32%
mWHO-BORR, 14%, 21%, 32%
Median irPFS, 4.6 m, 5.5 m, 5.7 m
Median PFS, 4.2 m, 4.1 m, 5.1 m
Median OS, 8.3 m, 9.7 m, 12.2 m
G3-4
6%
20%
15%
NCT 00527735 [49]
II R 1: 1: 1
N = 130
1. Placebo+Pacl+Carbo
2. Concurrent Ipi+Pacl+Carbo
3. Phased Ipi+Pacl+Carbo
irBORR, 53%, 49%, 71%
mWHO-BORR, 49%, 33%, 57%
Median irPFS, 5.3 m, 5.7 m, 6.4 m
Median PFS, 5.2 m, 3.9 m, 5.2 m
Median OS, 9.9 m, 9.1 m,12.9 m
G3-4
9%
21%
17%
NCT01285609 [50]
III R 1: 1
N = 749
1. Phased Placebo+Pacl+Carbo
2. Phased Ipi+Pacl+Carbo
BORR, 47%, 44%
Median PFS, 5.6 m, 5.6 m
Median OS, 12.4 m, 13.4 m
G3-4
35%
51%
CA184-156 [51]
III R 1, 1
N = 954
1. Phased Placebo+Etop+Plat
2. Phased Ipi+Etop+Plat
BORR, 62%, 62%
Median PFS, 4.4 m, 4.6 m
Median OS, 10.9 m, 11.0 m
G3-5
11%
22%
IMpower131 [52]
III R 1: 1
N = 578
1. Atezo+Nab-Pacl+Carbo
2. Nab-Pacl+Carbo
ORR, 49%, 41%
Median PFS, 6.3 m, 5.6 m
Median OS, 14.0 m, 13.9 m
G3-5
67%
58%
IMpower133 [53]
III R 1: 1
N = 403
1. Atezo+Etop+Carbo
2. Placebo+Etop+Carbo
ORR, 60.2%, 64.4%
Median PFS, 5.2 m, 4.3 m
Median OS, 12.3 m, 10.3 m
G3-5
58.1%
57.6%
IMpower132 [54]
III R 1: 1
N = 403
1. Atezo+Pem+Plat
2. Placebo+Pem+Plat
ORR, 47%, 32%
Median PFS, 7.6 m, 5.2 m
Median OS,18.1 m, 13.6 m
G3-5
58.1%
57.6%
IMpower150 [55]
III R 1: 1
N = 692
1. Atezo+Bev+Pacl+Carbo
2. Bev+Pacl+Carbo
ORR, 63.5%, 48%
Median PFS, 8.3 m, 6.8 m
Median OS,19.2 m, 14.7 m
G3-5
58.5%
50%
Second-line and beyond
CheckMate 017 [56]
Squamous
III R 1: 1
N = 272
1. Nivo
2. Docetaxel
ORR, 20%, 9%
Median PFS, 3.5 m, 2.8 m
Median OS, 9.2 m, 6.0 m
1-year OS, 42%, 24%
G3-4
7%
55%
CheckMate 032 [57]
I/II NR, N = 216
1. Nivo 3 mg/kg
2. Nivo 1 mg/kg + Ipi 3 mg/kg
3. Nivo 3 mg/kg + Ipi 1 mg/kg.
ORR, 10%, 23%, 19%
Median PFS, 1.4 m, 2.6 m, 1.4 m
Median OS, 4·4 m, 7.7 m, 6.0 m
1-year OS, 33%, 43%, 35%
G3-4
13%
30%
19%
CheckMate 057 [58]
Nonsquamous
III R 1: 1
N = 582
1. Nivo
2. Docetaxel
ORR, 19%, 12%
Median PFS,2.3 m, 4.2 m
Median OS, 12.2 m, 9.4 m
1-year OS, 51%, 39%
G3-4
10%
54%
CheckMate 331 [59]
III R 1: 1
N = 569
1. Nivo
2. Topotecan/Amrubicin
ORR, 13.7%, 16.5%
Median PFS, 1.5 m, 3.8 m
Median OS, 7.5 m, 8.4 m
1-year OS, 37%, 34%
G3-4
4.7%
74%
KEYNOTE-010 [60]
PD-L1 ≥ 1%
II/III R 1: 1: 1
N = 1034
1. Pembro 2 mg/kg,
2. Pembro 10 mg/kg
3. Docetaxel 75 mg/m2
ORR, 18%, 18%, 9%
Median PFS, 3.9 m, 4.0 m, 4.0 m
Median OS, 10·4 m, 12.7 m, 8.5 m
1-year OS, 43.2%, 52.3%, 34.6%
G3-5
13%
16%
35%
KEYNOTE-028
KEYNOTE-158 [61]
Ib/II N = 83
Pembro
ORR, 19.3%
Median PFS, 2.0 m
Median OS, 7.7 m
1-year OS, 20.7%
G3-5
8%
POPLAR [62]
II R 1: 1
N = 287
1. Atezo
2. Docetaxel
ORR, 15%, 15%
Median PFS, 2.7 m, 3.0 m
Median OS, 12.6 m, 9.7 m
G3-4
11%
39%
IFCT-1603 [63]
II R 2: 1
N = 64
1. Atezo
2. Etop+Carbo/Topotecan
6-week ORR, 2.3%, 10%
Median PFS, 1.4 m, 4.3 m
Median OS, 11.4 m, 9.4 m
G3-4
4.2%
~ 35%
OAK [64]
III R 1: 1
N = 850
1. Atezo
2. Docetaxel
ORR, 14%, 13%
Median PFS, 2.8 m, 4.0 m
Median OS, 13.8 m, 9.6 m
G3-4
15%
43%
Maintenance
PACIFIC [65]
III R 2: 1
N = 709
1. CRT+Durva
2. CRT+Placebo
Median PFS, 17.2 m, 5.6 m
Median OS, NR, 28.7 m
1-year OS, 66.3%, 55.6%
G3-4
30.5% 26.1%
CheckMate 451
III R 1: 1: 1
N = 810
1. Nivo
2. Nivo+Ipi
3. Placebo
Maintenance nivo/nivo+ipi did not improve OS
NA
NCT02359019 [66]
II N = 45
Pembro
Median PFS, 1.4 m
Median OS, 9.6 m
~ 15%
Abbreviations: ED extensive-stage, LD limited-stage, R randomized, NR nonrandomized, ORR objective response rate, PFS progression-free survival, OS overall survival, TRAE treatment-related adverse events, G3-5 grade 3-5, Pacl paclitaxel, Carbo carboplatin, Ipi ipilimumab, Etop etoposide, Bev bevacizumab, ir immune related, BORR best overall response rate, mWHO modified WHO, Plat platinum, Nivo nivolumab, Pembro pembrolizumab, Atezo atezolizumab

First-line

First, Reck et al. conducted a randomized phase II trial to investigate ipilimumab in combination with chemotherapy in previously untreated patients with lung cancer (ED-SCLC, n = 130; NSCLC, n = 204) [48, 49]. For both the NSCLC and SCLC cohorts, phased ipilimumab with carboplatin and paclitaxel but not a concurrent regimen showed improved immune-related (ir) progression-free survival (PFS) and a numeric, though not statistically significant, increase in median overall survival (OS) over chemotherapy alone. Overall grade 3-4 (G3-4) immune-related adverse events (irAEs) were more common for phased ipilimumab. Later, the phase III CA184-156 study further evaluated the efficacy of phased ipilimumab with etoposide and platinum as the first-line regimen for ED-SCLC [51]. Disappointingly, phased ipilimumab did not significantly prolong PFS and OS over the placebo and produced more irAEs. The rate of treatment-related discontinuation was even higher (18% v 2%). Similar results were observed in patients with advanced squamous NSCLC [50]. Another phase II trial involving phased ipilimumab to first-line chemotherapy even reported a G3-5 irAE rate as high as 69.2% with 5 of 42 patients dying [67]. The unfavorable benefit to risk profile limits the first-line application of ipilimamab in lung cancer.
The addition of atezolizumab to chemotherapy as the first-line treatment significantly improved the ORR and PFS among patients with metastatic nonsquamous and squamous NSCLC in IMpower 131 and IMpower 132 [52, 54]. Among patients with metastatic nonsquamous NSCLC, atezolizumab plus bevacizumab and chemotherapy significantly improved PFS and OS in IMpower 150, which led to FDA approval of the combined regimen for the first-line treatment of nonsquamous NSCLC [55]. For ED-SCLC, the clinical efficacy and safety of atezolizumab plus chemotherapy was evaluated in IMpower133 [53]. The interim analysis of the intention-to-treat population in this phase III trial showed that the addition of atezolizumab to standard chemotherapy significantly prolonged OS and PFS over the placebo (median OS, 12.3 v 10.3 months and median PFS, 5.2 v 4.3 months) and increased the 1-year OS by 13.5% (51.7% v 38.2%). The safety profile was consistent with previous observations. In March 2019, this combined regimen was approved as a first-line therapy for ED-SCLC. Overall, atezolizumab in combination with chemotherapy as a first-line treatment could be a novel option for people with advanced NSCLC and ED-SCLC.
Pembrolizumab in combination with standard chemotherapy resulted in significantly prolonged OS and PFS compared with chemotherapy alone among patients with squamous and nonsquamous NSCLC in KEYNOTE-407 and KEYNOTE-189 [68, 69]. However, the attempt to use pembrolizumab in the first-line setting for SCLC is limited. Only the phase Ib PembroPlus trial indicated that the standard dose pembrolizumab could be safely combined with many chemotherapy regimens across 6 advanced solid tumors, including relapsed SCLC [70]. A randomized phase III trial, KEYNOTE-604, is underway to assess the clinical benefit of pembrolizumab in combination with etoposide and platinum as a first-line treatment for ES-SCLC (Table 2).
Table 2
Ongoing clinical trials of immune checkpoint blockades in SCLC
Study
Stage
Ph
Pts.
Design
Primary end point
NCT number
Primary completion
First-line
 KEYNOTE-604
ED
III
453
Pembro+EP v Placebo+EP
PFS, OS
NCT03066778
December 2019
 REACTION
ED
II
118
Pembro+EP v EP
6-month PFS
NCT02580994
August 2020
  NA
LD/ED
I
80
Pembro+EP ± RT
MTD
NCT02402920
July 31, 2023
  NA
LD
II / III
506
Atezo+EP+RT v EP+RT
PFS, OS
NCT03811002
May 2024
 CASPIAN
ED
III
988
Durva±Treme+EP v EP
PFS, OS
NCT030s43872
September 2019
 CLOVER
LDb
I
300
Durva±Treme+EP
DLT; AE
NCT03509012
April 2022
  PAVE
ED
II
55
Avelumab+EP
1- year PFS
NCT03568097
November 2020
Second-line and beyond
 BLOLUMA
LD/EDa
II
106
Nivo+Ipi
ORR
NCT03083691
March 2019
 MCC-19163
LD/ED
II
41
Nivo+Ipi+Ad.p53-DC vaccine
DCR
NCT03406715
April 2020
 CA209-9YT
ED
II
40
Nivo+Ipi
Teff/Treg
NCT03670056
September 2020
  NA
ED
II
29
Nivo+Gemcitabine
ORR
NCT03662074
September 2020
  AFT-17
LD/ED
II
98
Pembro v Topotecan
PFS
NCT02963090
May 2019
 KEYNOTE PN758
ED
I/ II
84
Pembro+Pegzilarginase
AE, ORR
NCT03371979
December 2020
  NA
LD/ED
II
80
Pembro+EP±RT
PD-L1
NCT02934503
October 2019
 ML39728
LD/ED
II
35
Atezo
ORR
NCT03262454
December 2019
 Winship3112-15
LD/ED
II
28
Durva±Treme+RT v RT
PFS, ORR
NCT02701400
January 2020
 PASSION
ED
II
135
SHR-1210+Apatinib
AE, ORR
NCT03417895
March 2019
Maintenance
 STIMULI
LD
II
260
Nivo+Ipi v Obsesrvation
PFS, OS
NCT02046733
October 2019
 CA209-840
ED
I/ II
21
Nivo+Ipi+RT
PFS
NCT03043599
April 2021
 ACHILES
LD
II
212
Atezo v Observation
2-year OS
NCT03540420
December 2023
  NA
LD
II
51
Durva+EP+RT
PFS
NCT03585998
June 2021
 ADRIATIC
LD
III
600
Durva±Treme v Placebo
PFS, OS
NCT03703297
June 2021
Abbreviations: ED extensive-stage, LD limited-stage, a including NSCLC, b including 3 other solid tumors, ORR objective response rate, PFS progression-free survival, OS overall survival, AE adverse event, Nivo nivolumab, Ipi ipilimumab, RT radiotherapy, Pem pembrolizumab, EP etoposide plus platinum, MTD maximum tolerated dose, Atezo atezolizumab, Durva durvalumab, Treme tremelimumab, DLT dose-limiting toxicities, NA not available

Second-line

As shown in Table 1, the second-line nivolumab monotherapy significantly improved ORR, PFS, and OS compared with docetaxel among patients with advanced squamous and nonsquamous NSCLC in CheckMate 017 and CheckMate 057 [56, 58]. The response rate to nivolumab monotherapy was approximately twice that of docetaxel (20% v 10%), and nivolumab extended OS by approximately 3 months over chemotherapy. For SCLC, in the nonrandomized cohort in CheckMate 032 [57], the ORR was 10% (10 of 98) and 23% (14 of 61), and the median OS was 4.4 and 7.7 months for patients receiving nivolumab 3 mg/kg and nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, respectively. One-year OS was 33% and 43% for the two groups, respectively. Based on this trial, nivolumab and nivolumab plus ipilimumab were added as category 2A recommendations to the NCCN guidelines [11]. In August 2018, under accelerated approval, FDA approved nivolumab for treating patients with relapsed SCLC after the failure of platinum-based chemotherapy and one or more other lines of treatment. Unfortunately, CheckMate 331, a randomized phase III trial, demonstrated that nivolumab was inferior to topotecan or amrubicin in improving ORR, PFS, and OS among patients with relapsed SCLC [59].
Based on KEYNOTE-010, pembrolizumab was approved as a second-line treatment for advanced NSCLC patients with PD-L1 expression on ≥ 1% of tumor cells [60]. The phase Ib KEYNOTE-028 trial showed favorable efficacy and tolerable safety of pembrolizumab in treating patients with relapsed ED-SCLC and PD-L1 expression on ≥ 1% of tumor and stromal cells [71]. Further, the phase II KEYNOTE-158 trial confirmed the beneficial role of pembrolizumab in treating SCLC [72]. The latest results of KEYNOTE-028 and KEYNOTE-158 from 2019 from the American Association for Cancer Research (AACR) showed that pembrolizumab produced a durable response with tolerable toxicity for advanced SCLC patients after ≥ 2 lines of prior therapy. The ORR was 19.6% (16 of 83), with 2 patients having a complete response (CR) and 14 having a partial response (PR). More than half (9 of 16) had a response duration of ≥ 18 months. The median PFS was 2.0 months, and the median OS was 7.7 months, with a 1-year OS rate of 20.7%. The toxicity was manageable, with a G3-5 AE incidence of 9% [61]. Despite the encouraging results of single-arm studies, large randomized controlled studies are needed.
Atezolizumab also significantly improved OS by 3 to 4 months over docetaxel in patients with previously treated NSCLC in POPLAR and OAK [62, 64]. Unfortunately, the randomized phase II IFCT-1603 trial indicated that atezolizumab was not superior to chemotherapy as a second-line treatment in SCLC [63].

Maintenance

Durvalumab, an anti-PD-L1 antibody, has significantly prolonged PFS by more than twofold compared with the placebo (17.2 v 5.6 months, HR = 0.51) among patients with unresectable stage III NSCLC without disease progression after concurrent chemoradiotherapy in PACIFIC [65]. For patients with SCLC, the clinical outcome of ICBs as a maintenance regimen is quite dissatisfactory. A single-group, phase II study showed that maintenance pembrolizumab did not prolong OS compared with historical chemotherapy after first-line chemotherapy in patients with ED-SCLC [66]. In November 2018, Bristol-Myers Squibb announced that the phase III CheckMate 451 trial did not meet the primary endpoint of OS with nivolumab and nivolumab plus ipilimumab versus the placebo as maintenance therapy in patients with ED-SCLC.
In conclusion, anti-PD-1/PD-L1 blockades have revolutionized the standard treatment for NSCLC across the first-line, second-line, and maintenance settings based on multiple large randomized controlled trials (Table 1). However, for SCLC, despite moderate benefit in phase I/II trials, ICBs did not outcompete traditional chemotherapy in large randomized trials. Only atezolizumab in combination with chemotherapy conferred a survival benefit in CheckMate 331. Table 2 lists the ongoing trials involving ICBs or combination regimens in SCLC.

Potential mechanisms underlying discrepant outcomes of ICBs between SCLC and NSCLC

SCLC, with a high TMB similar to that of NSCLC, is theoretically likely to respond to immunotherapy. However, the clinical outcome of ICBs in SCLC seems far less effective than in NSCLC. There is a need to define the underlying differences in immune regulation patterns between SCLC and NSCLC to further guide immunotherapy for SCLC.

Low PD-L1 expression in SCLC

PD-L1 expression reflects pre-existing antitumor immunity in the TME and has been associated with a clinical benefit from anti-PD-1/PD-L1 blockade across multiple cancer types, including NSCLC [7375]. PD-L1 expression in NSCLC is approximately 50–70% according to previous reports [55, 56, 58, 62, 64, 68, 69, 76]. In stark contrast to NSCLC, PD-L1 expression in SCLC has been reported to be relatively low in most studies, as listed in Table 3. Most studies showed less than 50% PD-L1 expression in SCLC. The reason for the disparity in PD-L1 expression in these findings is not well understood and may be explained by the differences in staining antibodies, scoring algorithms, tissue biopsy types (archival or fresh), and detection platforms (Table 3). Notably, substantial PD-L1 expression occurs on stroma cells, including tumor-infiltrating immune cells (TILs and TIMs), in SCLC and less in tumor cells. Some of these studies demonstrated a positive correlation between PD-L1 expression on TICs and a favorable clinical outcome for SCLC patients [41, 66, 77, 80, 81]. Emerging data indicated that infiltrating stroma cells, such as dendritic cells and macrophages, could have protumorigenic functions by shaping antitumor immunity and the response to immunotherapy [8486]. Perhaps PD-L1 expression in immune cells may play a exceptional role in the pathophysiological process in SCLC. Overall, the relatively low PD-L1 expression may be at least one reason that the efficacy of ICBs in SCLC is not as good as that in NSCLC.
Table 3
PD-L1 expression in SCLC
Author
Patientsc
IHC assay
Biopsy types
TC PD-L1 cutoff
Cell type
PD-L1 expression
Antonia et al. [57]
146
28-8/Dako
Archival or fresh
1%
5%
Tumor cell
16.4%
4.8%
Ott et al. [71]
145
22C3/Dako
Archival or fresh
1%
Tumor and
Stromal cell
31.7%
Hyun et al. [72]
92
22C3/Dako
Archival or fresh
1%
Tumor and stromal cell
45.7%
Inamura et al. [77]
74
E1 L3N/CST
Archival
5%
Tumor cell
18.9%
Yasuda et al. [78]
39
22C3/Dako
Archival
1%
Tumor cell
2.5%
Pujol et al. [63]
53
SP142/Ventana
Archival
1%
Tumor and stromal cell
2%
Gadgeel et al. [66]
30
20
22C3/Dako
Archival
1%
Tumor cell
Stromal cell
10%
40%
Schultheis et al. [79].
94d
5H1/Chen’s lab
E1 L3N/CST
Archival
1%
Tumor cell
Stromal cell
0.0%
TIMs, 18.5%
TILs, 48%
Berghoff et al. [41]
32
NA/Dako
Archival
5%
Tumor cell
Stromal cell
34.4%
TIMs, 28.1%
TILs, 25.0%
Yu et al. [80]
194e
SP142/Ventana
28-8/Dako
Archival
1%
Tumor cell
Stromal cell
16.5%
TILs,
56.2%, 44.8%
Kim et al. [81]
120
MAB1561/
R&D system
Archival
1%
Tumor cell
Stromal cell
14.2%
TILs, 23.3%
Komiya et al [82]
99
EPR1161/Abcam
Archival
5%
Tumor cell
82.8%
Ishii et al. [83]
102
NA/Abcam
Archival
5%
Tumor cell
71.6%
Abbreviations: c patients who are PD-L1 evaluable, d small cell carcinomas 94 patients (SCLC 61; extrapulmonary 33), e total 194 patients, LD-SCLC 98,ED-SCLC 96, IHC immunohistochemistry, TC tumor cell, CST cell signaling technology, TIMs tumor infiltration macrophage, TILs tumor-infiltrating lymphocytes, NA not available*

Downregulation of MHC molecules in SCLC

The downregulation of MHC molecules is an immune escape mechanism. In contrast to NSCLC cells, which readily express MHC class I molecules, most SCLC cell lines and tissues showed marked deficient expression of MHC class I molecules, thus directly preventing tumor cells from presenting neoantigens to CD8+ T cells in the lymph nodes and inhibiting CTL recognition in the TME [87, 88]. Additionally, due to the lack of IFN-γ inducible expression of class II transactivator (cIITA), MHC class II molecule expression is absent in SCLC cells and significantly lower in SCLC TILs compared with in NSCLC, resulting in reduced presentation of tumor neoantigens to CD4+ T cells. In addition, mainly induced by IFN-γ-producing T cells, MHC class II molecules also reflect a less immunogenic environment in SCLC compared with NSCLC [8991].

Immunosuppression induced by SCLC cells

The interaction between SCLC cells and the immune system seems to be more complicated than previously thought. Tumors may destroy the host immune system in a variety of ways. The functions of the immune system, including lymphocyte reactivity to lectins and cytokines and the production of cytokines, were even more impaired in patients with SCLC compared with patients with NSCLC. The peripheral blood lymphocytes (PBLs) of SCLC patients showed significantly lower proliferative responses to phytohemagglutinin and human recombinant interleukin 2 (IL-2) than those of the NSCLC and noncancer groups. The ability of PBL to produce lymphokines (IL-2 and macrophage-activating factor) was evidently impaired in the SCLC group but not in the NSCLC group [92]. In contrast to NSCLC, the suppression of cytokine secretion in SCLC was dependent on tumor load and could be improved upon the reduction of the tumor load [93]. In addition, Wang et al. found that IL-15 secreted by SCLC cells could contribute to local and systemic immune escape and poor prognosis by inhibiting CD4+ T cell proliferation and supporting Treg cell induction [47]. Moreover, CD47 mainly functions to inhibit the activation and phagocytosis of macrophages through CD47/SIRPα signaling[94]. The remarkable upregulation of CD47 on the SCLC cell surface may be another important immune escape mechanism by inhibiting the activation and phagocytosis of macrophages. Preliminary experiments suggested that blocking CD47 induced macrophage-mediated phagocytosis in SCLC cell lines and mouse xenograft models [95]. Dysregulation of the Fas/FasL signaling pathway in tumors by reducing Fas expression and increasing FasL expression could participate in tumor development and immune escape [96, 97]. Fas expression is markedly decreased and even completely lost in lung tumors. However, FasL expression is decreased in most NSCLCs but is upregulated in 91% of SCLCs. FasL overexpression in the context of Fas downregulation in SCLC confers the ability on SCLC cells of inducing paracrine killing of Fas-expressing cytotoxic T cells and inhibiting self-apoptosis. These results indicated that the heavy tumor burden in SCLC makes the immune system less functional than in NSCLC through IL-15 secretion and the CD47/SIRPα and Fas/FasL pathways.

Autocrine and paracrine regulation in SCLC

A distinguishing feature of SCLC is the substantial autocrine and paracrine stimulation by growth factor receptors and chemokine receptors. Bombesin/gastrin-releasing peptide (BN/GRP) is a relevant growth factor in SCLC, and its receptor in overexpressed in SCLC [98]. Compared with NSCLC, which does not secrete autocrine growth factor granulocyte chemotactic protein (GCP-2), SCLC constantly secretes GCP-2 and expresses its receptors, CXCR1 and CXCR2 [99]. Stem cell factor (SCF) and its receptor c-kit, expressed in 40–70% SCLC specimens, are important regulators of SCLC viability [100]. In contrast to NSCLC cell lines, SCLC cell lines do not express intercellular adhesion molecule-1 (ICAM-1), which is involved in the interaction with lymphokine-activated killer cells [101]. IL-8 acts as an autocrine and/or paracrine growth factor for lung cancer, mediated by CXCR1 and CXCR2 receptors on the tumor cell surface. However, NSCLC cells produce much higher levels of IL-8 than SCLC cells. As a result, IL-8 may attract neutrophils and induce the immune response in NSCLC but mainly promote autocrine growth in SCLC [102, 103]. Lopez-Gonzalez et al. found that NSCLC and SCLC cell lines had diverse expression levels of TGF-β and its receptor [104]. NSCLC synthesizes both TGF-β isoforms and TGF-β RII. In stark contrast, malignant transformation in SCLC alters the synthesis of TGF-β isoforms and TGF-β RII, thereby avoiding autocrine and paracrine growth inhibition by TGF-β in SCLC cells. Another study found that four to eight SCLC cell lines could constantly secrete biologically active TGF-β1 to suppress IL-2-dependent T cell growth, and a specific anti-TGF-β1 antibody blocked the immunosuppressive activity induced by the SCLC cell lines [105]. These studies suggested that SCLC cells could markedly promote self-growth via autocrine and paracrine regulation but had less immune stimulatory function in adjacent immune cells compared with NSCLC cells.
These comparative analyses of immune regulation patterns between SCLC and NSCLC partially explain the immunodeficiency observed in patients with SCLC and the worse immune response in SCLC patients. Detailed investigations are essential to determine the exact immune escape mechanisms in SCLC to support the development of immunotherapy for SCLC.

Future of immunotherapy for SCLC

As demonstrated above, there are many differences underlying immune regulation patterns between NSCLC and SCLC. We note that several potential strategies could possibly improve the clinical outcome of immunotherapy for SCLC.

Chemotherapy and radiotherapy prior to ICBs

The intrinsic nature of ED-SCLC is so aggressive that a great proportion of patients dropped out or discontinued without completing the whole treatment course [51]. As illustrated in part 3, SCLC cells can induce immunosuppression and promote autocrine and paracrine regulation. Immune suppression seems to be associated with tumor load and could improve upon the reduction of the tumor load [93]. Chemotherapy and radiotherapy can not only reduce the heavy tumor burden of SCLC and further reinvigorate immune function but can also elevate PD-L1 expression and tumor antigen presentation by MHC molecules, thus favoring subsequent immunotherapy [106, 107]. We speculate that chemotherapy and radiotherapy prior to ICBs could be effective in improving the clinical outcome of immunotherapy for SCLC.

Combination of anti-angiogenesis and ICBs

Angiogenesis is one of the hallmarks of cancer [108]. Anlotinib, an oral anti-angiogenesis tyrosine multikinase inhibitor, is an approved third-line or later therapy for advanced NSCLC according to the Chinese Food and Drug Administration (CFDA), based on the ALTER 0303 study [109]. ALTER 1202, a phase II trial, has also demonstrated that anlotinib significantly improves OS as a third-line or later treatment [110]. Combination therapy with antiangiogenic therapy is a new paradigm in the treatment of advanced-stage malignancies via normalizing tumor vasculature and increasing drug delivery [111, 112]. Vascular endothelial growth factor (VEGF), the major regulator in tumor angiogenesis, plays an important role in immune modulation by blocking dendritic cell (DC) differentiation, reducing T cell infiltration, inducing inhibitory immune cells (Tregs and MDSCs) and inhibiting T cell development [112, 113]. Theoretically, combining immunotherapy with antiangiogenic treatment may have a synergistic effect, enhance the efficacy of both. Atezolizumab in combination with bevacizumab and chemotherapy significantly improved OS among patients with metastatic nonsquamous NSCLC in IMpower 150 [55]. The combination of ICBs with antiangiogenic therapy could also be promising in SCLC.

Biomarker selection

Compared with NSCLC, PD-L1 expression is relatively low in SCLC. In NSCLC, multiple clinical trials have defined PD-L1 as a biomarker for pembrolizumab, and the FDA-approved IHC assay utilized cutoffs of a 50% tumor proportion score for first-line and 1% for second-line pembrolizumab therapy [114]. Although PD-L1 expression was associated with a survival benefit in SCLC as indicated in early phase studies [66, 71, 72], it is not a perfect biomarker in SCLC. TMB, another biomarker of immunotherapy, could also predict the clinical outcome of ICBs based on exploratory analyses of CheckMate 026 [39] and CheckMate 227 [76] in NSCLC. In SCLC, Hellmann et al. retrospectively evaluated 211 TMB-evaluable patients from the total nonrandomized and randomized cohorts of CheckMate 032 [33]. For patients receiving nivolumab monotherapy and nivolumab plus ipilizumab, the ORR was higher in patients with a high TMB (21.3% and 46.2%) than in patients with a medium (6.8% and 16.0%) or low (4.8% and 22.2%) TMB. The OS for SCLC patients with a high TMB treated with nivolumab plus ipilimumab (22 months) was nearly three times that (6–8 months) achieved by the historical topotecan chemotherapy [16, 17]. Additionally, there was no association between PD-L1 expression and TMB in lung cancer [33, 39, 115]. Patients receiving nivolumab with a high level of these two biomarkers had a higher ORR than those with only one of these factors in CheckMate 026 [39]. Up to now, there are no exact biomarkers for immunotherapy in SCLC, TMB seemed to be more important than PD-L1 expression and perhaps the combination of the two biomarkers will be more valuable for selecting patients who will benefit from ICBs.

Conclusion

SCLC is an aggressive cancer type with poor prognosis. Limitations in the current treatment choices for SCLC give impetus to the search for novel therapeutic approaches including immunotherapy. Despite the similar level and distribution of the TMB between NSCLC and SCLC, the clinical efficacy of ICBs for SCLC is far less pronounced than that for NSCLC. Most studies concerning ICBs in SCLC failed to elicit better clinical outcomes than traditional chemotherapy. The potential mechanisms involved may be (1) low PD-L1 expression in SCLC, (2) downregulation of MHC molecules in SCLC, (3) immunosuppression induced by SCLC cells, and (4) autocrine and paracrine regulation in SCLC. Nevertheless, ICBs also brought about promising clinical benefits in SCLC. Atezolizumab in combination with etoposide and carboplatin prolonged OS by 2 months over chemotherapy alone and has recently received FDA approval as the first-line treatment for SCLC.
How to improve the clinical efficacy of immunotherapy for SCLC is an essential issue. Based on the dysregulation of the immune system in SCLC, we expect that chemotherapy and radiotherapy prior to immunotherapy could be more effective in improving the clinical outcome of ICBs in patients with SCLC. The combination of ICBs with antiangiogenic therapy may be a novel option for SCLC. Last, TMB seemed to be a more valuable biomarker for the efficacy of ICBs in SCLC, and the combination of TMB and PD-L1 may be promising for selecting patients who benefit from immunotherapy in SCLC.

Acknowledgments

Not applicable
Not applicable
Not applicable

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMedCrossRef Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMedCrossRef
3.
Zurück zum Zitat American Cancer Society. Cancer facts & figures 2014. Atlanta, GA: American Cancer Society; 2014. American Cancer Society. Cancer facts & figures 2014. Atlanta, GA: American Cancer Society; 2014.
4.
Zurück zum Zitat Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smoking and lung cancer--relative risk estimates for the major histological types from a pooled analysis of case-control studies. Int J Cancer. 2012;131:1210–9.PubMedCrossRef Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smoking and lung cancer--relative risk estimates for the major histological types from a pooled analysis of case-control studies. Int J Cancer. 2012;131:1210–9.PubMedCrossRef
5.
6.
Zurück zum Zitat Khuder SA. Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis. Lung Cancer (Amsterdam, Netherlands). 2001;31:139–48.CrossRef Khuder SA. Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis. Lung Cancer (Amsterdam, Netherlands). 2001;31:139–48.CrossRef
8.
Zurück zum Zitat Peifer M, Fernandez-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44:1104–10.PubMedPubMedCentralCrossRef Peifer M, Fernandez-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44:1104–10.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Pleasance ED, Stephens PJ, O’Meara S, et al. A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature. 2010;463:184–90.PubMedCrossRef Pleasance ED, Stephens PJ, O’Meara S, et al. A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature. 2010;463:184–90.PubMedCrossRef
11.
Zurück zum Zitat National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Small cell lung cancer. 2019; Version 1. National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Small cell lung cancer. 2019; Version 1.
12.
Zurück zum Zitat Zatloukal P, Cardenal F, Szczesna A, et al. A multicenter international randomized phase III study comparing cisplatin in combination with irinotecan or etoposide in previously untreated small-cell lung cancer patients with extensive disease. Ann Oncol. 2010;21:1810–6.PubMedCrossRef Zatloukal P, Cardenal F, Szczesna A, et al. A multicenter international randomized phase III study comparing cisplatin in combination with irinotecan or etoposide in previously untreated small-cell lung cancer patients with extensive disease. Ann Oncol. 2010;21:1810–6.PubMedCrossRef
13.
Zurück zum Zitat Hanna N, Bunn PA Jr, Langer C, et al. Randomized phase III trial comparing irinotecan/cisplatin with etoposide/cisplatin in patients with previously untreated extensive-stage disease small-cell lung cancer. J Clin Oncol. 2006;24:2038–43.PubMedCrossRef Hanna N, Bunn PA Jr, Langer C, et al. Randomized phase III trial comparing irinotecan/cisplatin with etoposide/cisplatin in patients with previously untreated extensive-stage disease small-cell lung cancer. J Clin Oncol. 2006;24:2038–43.PubMedCrossRef
14.
Zurück zum Zitat Schmittel A, Sebastian M, Fischer von Weikersthal L, et al. A German multicenter, randomized phase III trial comparing irinotecan-carboplatin with etoposide-carboplatin as first-line therapy for extensive-disease small-cell lung cancer. Ann Oncol. 2011;22:1798–804.PubMedCrossRef Schmittel A, Sebastian M, Fischer von Weikersthal L, et al. A German multicenter, randomized phase III trial comparing irinotecan-carboplatin with etoposide-carboplatin as first-line therapy for extensive-disease small-cell lung cancer. Ann Oncol. 2011;22:1798–804.PubMedCrossRef
15.
Zurück zum Zitat Byers LA, Rudin CM. Small cell lung cancer: where do we go from here? Cancer. 2015;121:664–72.PubMedCrossRef Byers LA, Rudin CM. Small cell lung cancer: where do we go from here? Cancer. 2015;121:664–72.PubMedCrossRef
16.
Zurück zum Zitat von Pawel J, Jotte R, Spigel DR, et al. Randomized phase III trial of amrubicin versus topotecan as second-line treatment for patients with small-cell lung cancer. J Clin Oncol. 2014;32:4012–9.CrossRef von Pawel J, Jotte R, Spigel DR, et al. Randomized phase III trial of amrubicin versus topotecan as second-line treatment for patients with small-cell lung cancer. J Clin Oncol. 2014;32:4012–9.CrossRef
17.
Zurück zum Zitat O’Brien ME, Ciuleanu TE, Tsekov H, et al. Phase III trial comparing supportive care alone with supportive care with oral topotecan in patients with relapsed small-cell lung cancer. J Clin Oncol. 2006;24:5441–7.PubMedCrossRef O’Brien ME, Ciuleanu TE, Tsekov H, et al. Phase III trial comparing supportive care alone with supportive care with oral topotecan in patients with relapsed small-cell lung cancer. J Clin Oncol. 2006;24:5441–7.PubMedCrossRef
18.
Zurück zum Zitat Cheng S, Evans WK, Stys-Norman D, et al. Chemotherapy for relapsed small cell lung cancer: a systematic review and practice guideline. J Thorac Oncol. 2007;2:348–54.PubMedCrossRef Cheng S, Evans WK, Stys-Norman D, et al. Chemotherapy for relapsed small cell lung cancer: a systematic review and practice guideline. J Thorac Oncol. 2007;2:348–54.PubMedCrossRef
19.
Zurück zum Zitat Vesely MD, Kershaw MH, Schreiber RD, et al. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011;29:235–71.PubMedCrossRef Vesely MD, Kershaw MH, Schreiber RD, et al. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011;29:235–71.PubMedCrossRef
20.
Zurück zum Zitat Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–30.PubMedCrossRef Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–30.PubMedCrossRef
22.
Zurück zum Zitat Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10.CrossRefPubMed Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10.CrossRefPubMed
23.
Zurück zum Zitat Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood. 2018;131:58–67.PubMed Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood. 2018;131:58–67.PubMed
24.
Zurück zum Zitat Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedPubMedCentralCrossRef Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26.CrossRefPubMed Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26.CrossRefPubMed
27.
29.
Zurück zum Zitat Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.PubMedPubMedCentralCrossRef Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.PubMedCrossRef Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.PubMedCrossRef
33.
Zurück zum Zitat Hellmann MD, Callahan MK, Awad MM, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018;33:853–61.PubMedCrossRefPubMedCentral Hellmann MD, Callahan MK, Awad MM, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018;33:853–61.PubMedCrossRefPubMedCentral
34.
Zurück zum Zitat Coulie PG, Van den Eynde BJ, van der Bruggen P, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135–46.PubMedCrossRef Coulie PG, Van den Eynde BJ, van der Bruggen P, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135–46.PubMedCrossRef
35.
Zurück zum Zitat Chan TA, Yarchoan M, Jaffee E, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30:44–56.PubMedCrossRef Chan TA, Yarchoan M, Jaffee E, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30:44–56.PubMedCrossRef
36.
Zurück zum Zitat McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.PubMedPubMedCentralCrossRef McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.PubMedPubMedCentralCrossRef Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.PubMedPubMedCentralCrossRef Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wang W, Hodkinson P, McLaren F, et al. Histologic assessment of tumor-associated CD45(+) cell numbers is an independent predictor of prognosis in small cell lung cancer. Chest. 2013;143:146–51.PubMedCrossRef Wang W, Hodkinson P, McLaren F, et al. Histologic assessment of tumor-associated CD45(+) cell numbers is an independent predictor of prognosis in small cell lung cancer. Chest. 2013;143:146–51.PubMedCrossRef
41.
Zurück zum Zitat Berghoff AS, Ricken G, Wilhelm D, et al. Tumor infiltrating lymphocytes and PD-L1 expression in brain metastases of small cell lung cancer (SCLC). J Neurooncol. 2016;130:19–29.PubMedCrossRef Berghoff AS, Ricken G, Wilhelm D, et al. Tumor infiltrating lymphocytes and PD-L1 expression in brain metastases of small cell lung cancer (SCLC). J Neurooncol. 2016;130:19–29.PubMedCrossRef
42.
Zurück zum Zitat Eerola AK, Soini Y, Paakko P. A high number of tumor-infiltrating lymphocytes are associated with a small tumor size, low tumor stage, and a favorable prognosis in operated small cell lung carcinoma. Clin Cancer Res. 2000;6:1875–81.PubMed Eerola AK, Soini Y, Paakko P. A high number of tumor-infiltrating lymphocytes are associated with a small tumor size, low tumor stage, and a favorable prognosis in operated small cell lung carcinoma. Clin Cancer Res. 2000;6:1875–81.PubMed
43.
Zurück zum Zitat Studnicka M, Wirnsberger R, Neumann M, et al. Peripheral blood lymphocyte subsets and survival in small-cell lung cancer. Chest. 1994;105:1673–8.PubMedCrossRef Studnicka M, Wirnsberger R, Neumann M, et al. Peripheral blood lymphocyte subsets and survival in small-cell lung cancer. Chest. 1994;105:1673–8.PubMedCrossRef
44.
Zurück zum Zitat Xie D, Marks R, Zhang M, et al. Nomograms predict overall survival for patients with small-cell lung cancer incorporating pretreatment peripheral blood markers. J Thorac Oncol. 2015;10:1213–20.PubMedCrossRef Xie D, Marks R, Zhang M, et al. Nomograms predict overall survival for patients with small-cell lung cancer incorporating pretreatment peripheral blood markers. J Thorac Oncol. 2015;10:1213–20.PubMedCrossRef
45.
Zurück zum Zitat Tian T, Gu X, Zhang B, et al. Increased circulating CD14(+)HLA-DR-/low myeloid-derived suppressor cells are associated with poor prognosis in patients with small-cell lung cancer. Cancer Biomarkers. 2015;15:425–32.PubMedCrossRef Tian T, Gu X, Zhang B, et al. Increased circulating CD14(+)HLA-DR-/low myeloid-derived suppressor cells are associated with poor prognosis in patients with small-cell lung cancer. Cancer Biomarkers. 2015;15:425–32.PubMedCrossRef
46.
Zurück zum Zitat Zhou T, Zhan J, Hong S, et al. Ratio of C-reactive protein/albumin is an inflammatory prognostic score for predicting overall survival of patients with small-cell lung cancer. Scientific reports. 2015;5:10481.PubMedPubMedCentralCrossRef Zhou T, Zhan J, Hong S, et al. Ratio of C-reactive protein/albumin is an inflammatory prognostic score for predicting overall survival of patients with small-cell lung cancer. Scientific reports. 2015;5:10481.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Wang W, Hodkinson P, McLaren F, et al. Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3+ cells in tumour infiltrate. Int J Cancer. 2012;131:E928–37.PubMedCrossRef Wang W, Hodkinson P, McLaren F, et al. Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3+ cells in tumour infiltrate. Int J Cancer. 2012;131:E928–37.PubMedCrossRef
48.
Zurück zum Zitat Lynch TJ, Bondarenko I, Luft A, 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.PubMedCrossRef Lynch TJ, Bondarenko I, Luft A, 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.PubMedCrossRef
49.
Zurück zum Zitat Reck M, Bondarenko I, Luft A, et al. 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;24:75–83.CrossRefPubMed Reck M, Bondarenko I, Luft A, et al. 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;24:75–83.CrossRefPubMed
50.
Zurück zum Zitat Govindan R, Szczesna A, Ahn MJ, et al. Phase III trial of ipilimumab combined with paclitaxel and carboplatin in advanced squamous non-small-cell lung cancer. J Clin Oncol. 2017;35:3449–57.PubMedCrossRef Govindan R, Szczesna A, Ahn MJ, et al. Phase III trial of ipilimumab combined with paclitaxel and carboplatin in advanced squamous non-small-cell lung cancer. J Clin Oncol. 2017;35:3449–57.PubMedCrossRef
51.
Zurück zum Zitat Reck M, Luft A, Szczesna A, et al. Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer. J Clin Oncol. 2016;34:3740–8.PubMedCrossRef Reck M, Luft A, Szczesna A, et al. Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer. J Clin Oncol. 2016;34:3740–8.PubMedCrossRef
52.
Zurück zum Zitat Jotte RMCF, Vynnychenko I, et al. IMpower131: Primary PFS and safety analysis of a randomized phase III study of atezolizumab + carboplatin +paclitaxel or nab-paclitaxel vs carboplatin + nab-paclitaxel as 1 L therapy in advanced squamous NSCLC. J Clin Oncol. 2018;36(18_suppl):LBA9000.CrossRef Jotte RMCF, Vynnychenko I, et al. IMpower131: Primary PFS and safety analysis of a randomized phase III study of atezolizumab + carboplatin +paclitaxel or nab-paclitaxel vs carboplatin + nab-paclitaxel as 1 L therapy in advanced squamous NSCLC. J Clin Oncol. 2018;36(18_suppl):LBA9000.CrossRef
53.
Zurück zum Zitat Horn L, Mansfield AS, Szczesna A, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 2018;379:2220–9.PubMedCrossRef Horn L, Mansfield AS, Szczesna A, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 2018;379:2220–9.PubMedCrossRef
54.
Zurück zum Zitat Barlesi F, MN MC, et al. IMpower 132: Atezolizumab in combination with pemetrexed and platinum-based chemotherapy in advanced nonsquamous NSCLC: WCLC; 2018. Barlesi F, MN MC, et al. IMpower 132: Atezolizumab in combination with pemetrexed and platinum-based chemotherapy in advanced nonsquamous NSCLC: WCLC; 2018.
55.
Zurück zum Zitat Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–301.PubMedCrossRef Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–301.PubMedCrossRef
56.
Zurück zum Zitat Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123–35.PubMedPubMedCentralCrossRef Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123–35.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Antonia SJ, Lopez-Martin JA, Bendell J, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:883–95.PubMedCrossRef Antonia SJ, Lopez-Martin JA, Bendell J, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:883–95.PubMedCrossRef
58.
Zurück zum Zitat Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–39.PubMedPubMedCentralCrossRef Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–39.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Reck MVD, Ciuleanu T, et al. Efficacy and safety of nivolumab (nivo) monotherapy versus chemotherapy (chemo) in recurrent small cell lung cancer (SCLC): Results from CheckMate 331: ESMO; 2018. Reck MVD, Ciuleanu T, et al. Efficacy and safety of nivolumab (nivo) monotherapy versus chemotherapy (chemo) in recurrent small cell lung cancer (SCLC): Results from CheckMate 331: ESMO; 2018.
60.
Zurück zum Zitat Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet (London, England). 2016;387:1540–50.PubMedCrossRef Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet (London, England). 2016;387:1540–50.PubMedCrossRef
61.
Zurück zum Zitat Chung HC P-PS, Lopez-Martin J,et al. Pembrolizumab after two or more lines of prior therapy in patients with advanced small-cell lung cancer (SCLC):results from the KEYNOTE-028 and KEYNOTE-158 studies. AACR 2019; abstract CT073. Chung HC P-PS, Lopez-Martin J,et al. Pembrolizumab after two or more lines of prior therapy in patients with advanced small-cell lung cancer (SCLC):results from the KEYNOTE-028 and KEYNOTE-158 studies. AACR 2019; abstract CT073.
62.
Zurück zum Zitat Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet (London, England). 2016;387:1837–46.CrossRef Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet (London, England). 2016;387:1837–46.CrossRef
63.
Zurück zum Zitat Pujol JL, Greillier L, Audigier-Valette C, et al. A randomized non-comparative phase 2 study of anti-programmed cell death-ligand 1 atezolizumab or chemotherapy as second-line therapy in patients with small cell lung cancer: results from the IFCT-1603 trial. J Thorac Oncol. 2019. Pujol JL, Greillier L, Audigier-Valette C, et al. A randomized non-comparative phase 2 study of anti-programmed cell death-ligand 1 atezolizumab or chemotherapy as second-line therapy in patients with small cell lung cancer: results from the IFCT-1603 trial. J Thorac Oncol. 2019.
64.
Zurück zum Zitat Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet (London, England). 2017;389:255–65.CrossRef Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet (London, England). 2017;389:255–65.CrossRef
65.
Zurück zum Zitat Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379:2342–50.PubMedCrossRef Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379:2342–50.PubMedCrossRef
66.
Zurück zum Zitat Gadgeel SM, Pennell NA, Fidler MJ, et al. Phase II study of maintenance pembrolizumab in patients with extensive-stage small cell lung cancer (SCLC). J Thorac Oncol. 2018;13:1393–9.PubMedCrossRefPubMedCentral Gadgeel SM, Pennell NA, Fidler MJ, et al. Phase II study of maintenance pembrolizumab in patients with extensive-stage small cell lung cancer (SCLC). J Thorac Oncol. 2018;13:1393–9.PubMedCrossRefPubMedCentral
67.
Zurück zum Zitat Arriola E, Wheater M, Galea I, et al. Outcome and biomarker analysis from a multicenter phase 2 study of ipilimumab in combination with carboplatin and etoposide as first-line therapy for extensive-stage SCLC. J Thorac Oncol. 2016;11:1511–21.PubMedPubMedCentralCrossRef Arriola E, Wheater M, Galea I, et al. Outcome and biomarker analysis from a multicenter phase 2 study of ipilimumab in combination with carboplatin and etoposide as first-line therapy for extensive-stage SCLC. J Thorac Oncol. 2016;11:1511–21.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379:2040–51.PubMedCrossRef Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379:2040–51.PubMedCrossRef
69.
Zurück zum Zitat Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.PubMedCrossRef Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.PubMedCrossRef
70.
Zurück zum Zitat Weiss GJ, Waypa J, Blaydorn L, et al. A phase Ib study of pembrolizumab plus chemotherapy in patients with advanced cancer (PembroPlus). Br J Cancer. 2017;117:33–40.PubMedPubMedCentralCrossRef Weiss GJ, Waypa J, Blaydorn L, et al. A phase Ib study of pembrolizumab plus chemotherapy in patients with advanced cancer (PembroPlus). Br J Cancer. 2017;117:33–40.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Ott PA, Elez E, Hiret S, et al. Pembrolizumab in patients with extensive-stage small-cell lung cancer: results from the phase Ib KEYNOTE-028 Study. J Clin Oncol. 2017;35:3823–9.PubMedCrossRef Ott PA, Elez E, Hiret S, et al. Pembrolizumab in patients with extensive-stage small-cell lung cancer: results from the phase Ib KEYNOTE-028 Study. J Clin Oncol. 2017;35:3823–9.PubMedCrossRef
72.
Zurück zum Zitat Chung HCL-MJ, Kao SCH, Miller WH, Ros W, Gao B. Phase 2 study of pembrolizumab in advanced small-cell lung cancer (SCLC): KEYNOTE-158. J Clin Oncol. 2018;15(36):8506.CrossRef Chung HCL-MJ, Kao SCH, Miller WH, Ros W, Gao B. Phase 2 study of pembrolizumab in advanced small-cell lung cancer (SCLC): KEYNOTE-158. J Clin Oncol. 2018;15(36):8506.CrossRef
73.
Zurück zum Zitat Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–56.PubMedCrossRef Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–56.PubMedCrossRef
74.
Zurück zum Zitat Sacher AG, Gandhi L. biomarkers for the clinical use of PD-1/PD-L1 inhibitors in non-small-cell lung cancer: a review. JAMA Oncol. 2016;2:1217–22.PubMedCrossRef Sacher AG, Gandhi L. biomarkers for the clinical use of PD-1/PD-L1 inhibitors in non-small-cell lung cancer: a review. JAMA Oncol. 2016;2:1217–22.PubMedCrossRef
75.
Zurück zum Zitat Kerr KM, Tsao MS, Nicholson AG, et al. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10:985–9.PubMedCrossRef Kerr KM, Tsao MS, Nicholson AG, et al. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10:985–9.PubMedCrossRef
76.
Zurück zum Zitat Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378:2093–104.PubMedCrossRefPubMedCentral Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378:2093–104.PubMedCrossRefPubMedCentral
77.
Zurück zum Zitat Inamura K, Yokouchi Y, Kobayashi M, et al. Relationship of tumor PD-L1 (CD274) expression with lower mortality in lung high-grade neuroendocrine tumor. Cancer Med. 2017;6:2347–56.PubMedPubMedCentralCrossRef Inamura K, Yokouchi Y, Kobayashi M, et al. Relationship of tumor PD-L1 (CD274) expression with lower mortality in lung high-grade neuroendocrine tumor. Cancer Med. 2017;6:2347–56.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Yasuda Y, Ozasa H, Kim YH. PD-L1 expression in small cell lung cancer. J Thorac Oncol. 2018;13:e40–1.PubMedCrossRef Yasuda Y, Ozasa H, Kim YH. PD-L1 expression in small cell lung cancer. J Thorac Oncol. 2018;13:e40–1.PubMedCrossRef
79.
Zurück zum Zitat Schultheis AM, Scheel AH, Ozretic L, et al. PD-L1 expression in small cell neuroendocrine carcinomas. Eur J Cancer. 2015;51:421–6.PubMedCrossRef Schultheis AM, Scheel AH, Ozretic L, et al. PD-L1 expression in small cell neuroendocrine carcinomas. Eur J Cancer. 2015;51:421–6.PubMedCrossRef
80.
Zurück zum Zitat Yu H, Batenchuk C, Badzio A, et al. PD-L1 Expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J Thorac Oncol. 2017;12:110–20.PubMedCrossRef Yu H, Batenchuk C, Badzio A, et al. PD-L1 Expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J Thorac Oncol. 2017;12:110–20.PubMedCrossRef
81.
Zurück zum Zitat Kim HS, Lee JH, Nam SJ, et al. Association of PD-L1 expression with tumor-infiltrating immune cells and mutation burden in high-grade neuroendocrine carcinoma of the lung. J Thorac Oncol. 2018;13:636–48.PubMedCrossRef Kim HS, Lee JH, Nam SJ, et al. Association of PD-L1 expression with tumor-infiltrating immune cells and mutation burden in high-grade neuroendocrine carcinoma of the lung. J Thorac Oncol. 2018;13:636–48.PubMedCrossRef
82.
Zurück zum Zitat Ishii H, Azuma K, Kawahara A, et al. Significance of programmed cell death-ligand 1 expression and its association with survival in patients with small cell lung cancer. J Thorac Oncol. 2015;10:426–30.PubMedCrossRef Ishii H, Azuma K, Kawahara A, et al. Significance of programmed cell death-ligand 1 expression and its association with survival in patients with small cell lung cancer. J Thorac Oncol. 2015;10:426–30.PubMedCrossRef
83.
Zurück zum Zitat Komiya T, Madan R. PD-L1 expression in small cell lung cancer. Eur J Cancer. 2015;51:1853–5.PubMedCrossRef Komiya T, Madan R. PD-L1 expression in small cell lung cancer. Eur J Cancer. 2015;51:1853–5.PubMedCrossRef
84.
Zurück zum Zitat Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15:669–82.PubMedCrossRef Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15:669–82.PubMedCrossRef
85.
Zurück zum Zitat Tang H, Liang Y, Anders RA, et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018;128:580–8.PubMedPubMedCentralCrossRef Tang H, Liang Y, Anders RA, et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018;128:580–8.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Lin H, Wei S, Hurt EM, et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest. 2018;128:805–15.PubMedPubMedCentralCrossRef Lin H, Wei S, Hurt EM, et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest. 2018;128:805–15.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Doyle A. Markedly decreased expression of class I histocompatibility antigens, protein, and mRNA in human small-cell lung cancer. J Exp Med. 1985;161:1135–51.PubMedCrossRef Doyle A. Markedly decreased expression of class I histocompatibility antigens, protein, and mRNA in human small-cell lung cancer. J Exp Med. 1985;161:1135–51.PubMedCrossRef
88.
Zurück zum Zitat Traversari C, Meazza R, Coppolecchia M, et al. IFN-gamma gene transfer restores HLA-class I expression and MAGE-3 antigen presentation to CTL in HLA-deficient small cell lung cancer. Gene Therapy. 1997;4:1029–35.PubMedCrossRef Traversari C, Meazza R, Coppolecchia M, et al. IFN-gamma gene transfer restores HLA-class I expression and MAGE-3 antigen presentation to CTL in HLA-deficient small cell lung cancer. Gene Therapy. 1997;4:1029–35.PubMedCrossRef
89.
Zurück zum Zitat He Y, Rozeboom L, Rivard CJ, et al. MHC class II expression in lung cancer. Lung Cancer (Amsterdam, Netherlands). 2017;112:75–80.CrossRef He Y, Rozeboom L, Rivard CJ, et al. MHC class II expression in lung cancer. Lung Cancer (Amsterdam, Netherlands). 2017;112:75–80.CrossRef
90.
Zurück zum Zitat Yazawa T, Ito T, Kamma H, et al. Complicated mechanisms of class II transactivator transcription deficiency in small cell lung cancer and neuroblastoma. Am J Pathol. 2002;161:291–300.PubMedPubMedCentralCrossRef Yazawa T, Ito T, Kamma H, et al. Complicated mechanisms of class II transactivator transcription deficiency in small cell lung cancer and neuroblastoma. Am J Pathol. 2002;161:291–300.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Yazawa T, Kamma H, Fujiwara M, et al. Lack of class II transactivator causes severe deficiency of HLA-DR expression in small cell lung cancer. J Pathol. 1999;187:191–9.PubMedCrossRef Yazawa T, Kamma H, Fujiwara M, et al. Lack of class II transactivator causes severe deficiency of HLA-DR expression in small cell lung cancer. J Pathol. 1999;187:191–9.PubMedCrossRef
92.
Zurück zum Zitat Masuno T, Ikeda T, Yokota S, et al. Immunoregulatory T-lymphocyte functions in patients with small cell lung cancer. Cancer Res. 1986;46:4195–9.PubMed Masuno T, Ikeda T, Yokota S, et al. Immunoregulatory T-lymphocyte functions in patients with small cell lung cancer. Cancer Res. 1986;46:4195–9.PubMed
93.
Zurück zum Zitat Fischer JR, Schindel M, Stein N, et al. Selective suppression of cytokine secretion in patients with small-cell lung cancer. Ann Oncol. 1995;6:921–6.PubMedCrossRef Fischer JR, Schindel M, Stein N, et al. Selective suppression of cytokine secretion in patients with small-cell lung cancer. Ann Oncol. 1995;6:921–6.PubMedCrossRef
94.
Zurück zum Zitat Matozaki T, Murata Y, Okazawa H, et al. Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009;19:72–80.PubMedCrossRef Matozaki T, Murata Y, Okazawa H, et al. Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009;19:72–80.PubMedCrossRef
95.
Zurück zum Zitat Weiskopf K, Jahchan NS, Schnorr PJ, et al. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer. J Clin Invest. 2016;126:2610–20.PubMedPubMedCentralCrossRef Weiskopf K, Jahchan NS, Schnorr PJ, et al. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer. J Clin Invest. 2016;126:2610–20.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Viard-Leveugle I, Veyrenc S, French LE, et al. Frequent loss of Fas expression and function in human lung tumours with overexpression of FasL in small cell lung carcinoma. J Pathol. 2003;201:268–77.PubMedCrossRef Viard-Leveugle I, Veyrenc S, French LE, et al. Frequent loss of Fas expression and function in human lung tumours with overexpression of FasL in small cell lung carcinoma. J Pathol. 2003;201:268–77.PubMedCrossRef
97.
Zurück zum Zitat Hopkins-Donaldson S, Ziegler A, Kurtz S, et al. Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation. Cell Death Differ. 2003;10:356–64.PubMedCrossRef Hopkins-Donaldson S, Ziegler A, Kurtz S, et al. Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation. Cell Death Differ. 2003;10:356–64.PubMedCrossRef
98.
Zurück zum Zitat Zhou J, Chen J, Mokotoff M, et al. Bombesin/gastrin-releasing peptide receptor: a potential target for antibody-mediated therapy of small cell lung cancer. Clin Cancer Res. 2003;9:4953–60.PubMed Zhou J, Chen J, Mokotoff M, et al. Bombesin/gastrin-releasing peptide receptor: a potential target for antibody-mediated therapy of small cell lung cancer. Clin Cancer Res. 2003;9:4953–60.PubMed
99.
Zurück zum Zitat Zhu YM, Bagstaff SM, Woll PJ. Production and upregulation of granulocyte chemotactic protein-2/CXCL6 by IL-1beta and hypoxia in small cell lung cancer. Br J Cancer. 2006;94:1936–41.PubMedPubMedCentralCrossRef Zhu YM, Bagstaff SM, Woll PJ. Production and upregulation of granulocyte chemotactic protein-2/CXCL6 by IL-1beta and hypoxia in small cell lung cancer. Br J Cancer. 2006;94:1936–41.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Kijima T, Maulik G, Ma PC, et al. Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res. 2002;62:6304–11.PubMed Kijima T, Maulik G, Ma PC, et al. Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res. 2002;62:6304–11.PubMed
101.
Zurück zum Zitat Melis M, Spatafora M, Melodia A, et al. ICAM-1 expression by lung cancer cell lines: effects of upregulation by cytokines on the interaction with LAK cells. Eur Respir J. 1996;9:1831–8.PubMedCrossRef Melis M, Spatafora M, Melodia A, et al. ICAM-1 expression by lung cancer cell lines: effects of upregulation by cytokines on the interaction with LAK cells. Eur Respir J. 1996;9:1831–8.PubMedCrossRef
102.
103.
Zurück zum Zitat Shibakura M, Niiya K, Kiguchi T, et al. Induction of IL-8 and monoclyte chemoattractant protein-1 by doxorubicin in human small cell lung carcinoma cells. Int J Cancer. 2003;103:380–6.PubMedCrossRef Shibakura M, Niiya K, Kiguchi T, et al. Induction of IL-8 and monoclyte chemoattractant protein-1 by doxorubicin in human small cell lung carcinoma cells. Int J Cancer. 2003;103:380–6.PubMedCrossRef
104.
Zurück zum Zitat Lopez-Gonzalez JS, Aguilar-Cazares D, Prado-Garcia H, et al. Lack of correlation between growth inhibition by TGF-beta and the percentage of cells expressing type II TGF-beta receptor in human non-small cell lung carcinoma cell lines. Lung Cancer (Amsterdam, Netherlands). 2002;38:149–58.CrossRef Lopez-Gonzalez JS, Aguilar-Cazares D, Prado-Garcia H, et al. Lack of correlation between growth inhibition by TGF-beta and the percentage of cells expressing type II TGF-beta receptor in human non-small cell lung carcinoma cell lines. Lung Cancer (Amsterdam, Netherlands). 2002;38:149–58.CrossRef
105.
Zurück zum Zitat Fischer JR, Darjes H, Lahm H, et al. Constitutive secretion of bioactive transforming growth factor beta 1 by small cell lung cancer cell lines. Eur J Cancer. 1994;30a:2125–9.PubMedCrossRef Fischer JR, Darjes H, Lahm H, et al. Constitutive secretion of bioactive transforming growth factor beta 1 by small cell lung cancer cell lines. Eur J Cancer. 1994;30a:2125–9.PubMedCrossRef
106.
Zurück zum Zitat Ko EC, Raben D, Formenti SC. The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer. Clin Cancer Res. 2018;24:5792–806.PubMedCrossRef Ko EC, Raben D, Formenti SC. The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer. Clin Cancer Res. 2018;24:5792–806.PubMedCrossRef
107.
Zurück zum Zitat Zitvogel L, Apetoh L, Ghiringhelli F, et al. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73.PubMedCrossRef Zitvogel L, Apetoh L, Ghiringhelli F, et al. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73.PubMedCrossRef
109.
Zurück zum Zitat Han B, Li K, Wang Q, et al. Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: the ALTER 0303 phase 3 randomized clinical trial. JAMA Oncol. 2018;4:1569–75.PubMedPubMedCentralCrossRef Han B, Li K, Wang Q, et al. Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: the ALTER 0303 phase 3 randomized clinical trial. JAMA Oncol. 2018;4:1569–75.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Cheng Y WQ, Li K, Shi J, Wu L, Han B, et al. Anlotinib as third-line or further-line treatment in relapsed SCLC: a multicentre, randomized, double-blind phase 2 trial. . WCLC 2018; OA 13.03. Cheng Y WQ, Li K, Shi J, Wu L, Han B, et al. Anlotinib as third-line or further-line treatment in relapsed SCLC: a multicentre, randomized, double-blind phase 2 trial. . WCLC 2018; OA 13.03.
111.
Zurück zum Zitat Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nature Med. 2001;7:987–9.PubMedCrossRef Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nature Med. 2001;7:987–9.PubMedCrossRef
112.
Zurück zum Zitat Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579.PubMedCrossRef Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579.PubMedCrossRef
113.
Zurück zum Zitat Manegold C, Dingemans AC, Gray JE, et al. The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J Thorac Oncol. 2017;12:194–207.PubMedCrossRef Manegold C, Dingemans AC, Gray JE, et al. The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J Thorac Oncol. 2017;12:194–207.PubMedCrossRef
114.
Zurück zum Zitat National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Non-small cell lung cancer. 2019; Version 3. National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Non-small cell lung cancer. 2019; Version 3.
115.
Zurück zum Zitat Hellmann MD, Nathanson T, Rizvi H, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33:843–852 e844.PubMedPubMedCentralCrossRef Hellmann MD, Nathanson T, Rizvi H, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33:843–852 e844.PubMedPubMedCentralCrossRef
Metadaten
Titel
Potential immune escape mechanisms underlying the distinct clinical outcome of immune checkpoint blockades in small cell lung cancer
verfasst von
Yaru Tian
Xiaoyang Zhai
Anqin Han
Hui Zhu
Jinming Yu
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2019
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0753-2

Weitere Artikel der Ausgabe 1/2019

Journal of Hematology & Oncology 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.