Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Review

Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors

verfasst von: Ming Yi, Dechao Jiao, Hanxiao Xu, Qian Liu, Weiheng Zhao, Xinwei Han, Kongming Wu

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

Programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) is a negative modulatory signaling pathway for activation of T cell. It is acknowledged that PD-1/PD-L1 axis plays a crucial role in the progression of tumor by altering status of immune surveillance. As one of the most promising immune therapy strategies, PD-1/PD-L1 inhibitor is a breakthrough for the therapy of some refractory tumors. However, response rate of PD-1/PD-L1 inhibitors in overall patients is unsatisfactory, which limits the application in clinical practice. Therefore, biomarkers which could effectively predict the efficacy of PD-1/PD-L1 inhibitors are crucial for patient selection. Biomarkers reflecting tumor immune microenvironment and tumor cell intrinsic features, such as PD-L1 expression, density of tumor infiltrating lymphocyte (TIL), tumor mutational burden, and mismatch-repair (MMR) deficiency, have been noticed to associate with treatment effect of anti-PD-1/anti-PD-L1 therapy. Furthermore, gut microbiota, circulating biomarkers, and patient previous history have been found as valuable predictors as well. Therefore establishing a comprehensive assessment framework involving multiple biomarkers would be meaningful to interrogate tumor immune landscape and select sensitive patients.
Hinweise
Ming Yi and Dechao Jiao contributed equally to this work.
Abkürzungen
ALK
Anaplastic lymphoma kinase
APOBEC3
Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3
COPD
Chronic obstructive pulmonary disease
CTLA-4
Cytotoxic T-lymphocyte-associated protein 4
DC
Dendritic cell
dMMR
Mismatch repair deficiency
EGFR
Epidermal growth factor receptor
EMT
epithelial-to-mesenchymal transition
HR
hazard ratio
IC
Immune cell
ICAM-1
Intercellular cell adhesion molecule-1
ICOS
Inducible T cell costimulator
ITIM
Immune receptor tyrosine based inhibitory motif
ITSM
Immune receptor tyrosine-based switch motif
KRAS
Kirsten rat sarcoma viral oncogene homology
LDH
Lactate dehydrogenase
mEGFR
EGFR activating mutation
MLH1
MutL homolog 1
MMR
Mismatch repair
MSH2
MutS protein homolog 2
MSH6
MutS homolog 6
NSCLC
Non-small-cell-lung cancer
OS
Overcall survival
PD-1/PD-L1
Programmed cell death protein 1/programmed cell death 1 ligand 1
PMS2
PMS1 homolog 2
POLD1
Polymerase δ1
POLE
polymerase ε
PRL
Prolactin;
SCFA
Short chain fatty acid
SHP1/2
Src homology 2 domain containing phosphatases 1/2
sPD-L1
soluble PD-L1
TC
Tumor cell
TCM
Central memory T cell
TGF-β
Transforming growth factor β
TMB
tumor mutational burden
TNF-α
Tumor necrosis factor-α
VEGF
Vascular endothelial growth factor

Background

Novel cancer immunotherapy is the most promising cancer treatment strategy, mainly including chimeric antigen receptor T cell, bispecific antibodies and immune checkpoint inhibitors [14]. Programmed cell death protein 1/programmed cell death-ligand 1 (PD-1/PD-L1) axis is a vital immune checkpoint signaling pathway which could downregulate magnitude of inflammation response and maintain immune homeostasis [5]. Immune receptor tyrosine based inhibitory motif (ITIM) and immune receptor tyrosine-based switch motif (ITSM) are core structures of PD-1, which transduct extracellular signal and recruit Src homology 2 domain containing phosphatases 1/2 (SHP1/2) within the cell [6]. PD-1/PD-L1 axis impairs activation of T cell by inhibiting Ras-Raf-MEK-ERK and PI3K-AKT signaling pathways which are generally believed to promote proliferation and differentiation of T cell [7]. The inhibitory regulation of PD-1/PD-L1 is usually compared to a brake for activation of T cell [8].
In the evolution of immunity, PD-1/PD-L1 axis is an indispensable pathway to maintain immune tolerance and prevent autoimmunity diseases [911]. However, PD-1/PD-L1 axis influence the balance between tumor immune surveillance and immune resistance as well [12, 13]. Elevated PD-L1 expression on tumor cell or tumor infiltrating lymphocyte (TIL) results in the exhaustion of T cell [14], thus the attenuated tumor-specific immunity promoting tumor progression [15].
Based on the mechanism mentioned above, PD-1/PD-L1 inhibitors block the negative regulatory signal pathways and unleash T cell from exhausted status [16]. Since first PD-1/PD-L1 inhibitor (pembrolizumab) was approved by Food and Drug Administration in 2014, many immune checkpoint inhibitors have been applied in clinical practice [17, 18]. PD-1/PD-L1 inhibitors show potent and durable anti-tumor effects, especially in some refractory tumors [4, 19, 20]. Even though the relatively low response rate limits the application in patients, PD-1/PD-L1 inhibitors attract extensive attention [2123].
In clinical practice, the primary problem for application of PD-1/PD-L1 inhibitors is the unsatisfactory response rate in overall patients. Therefore, patient selection should be implemented prior to PD-1/PD-L1 inhibitors therapy [24, 25]. Identifying predictive biomarkers to distinguish patients most likely to respond to immunotherapy from overall individuals would decrease treatment cost and avoid immune-related adverse events.
A possible mechanism of tumor immune escape is adaptive immune resistance, indicating the feedback that IFN-γ-induced upregulation of PD-1/PD-L1 axis could downregulate the cytokines and suppress the immune response in tumor microenvironment [26, 27]. Tumor regression induced by PD-1/PD-L1 inhibitors is influenced by some tumor microenvironment related factors such as PD-L1 status and pre-existing tumor infiltrating lymphocyte (TIL) [13, 26].

PD-L1 expression

Relationship between PD-L1 expression and therapeutic response rate

As the most widely adopted predictor, the role of PD-L1 expression has been investigated in many clinical trials (Table 1). Status of PD-L1 expression (positive/negative) is measured by proportion of PD-L1 expressing tumor cell (TC) and/or immune cell (IC). However, the conclusions from multiple trials are not consistent. Generally believed, high PD-L1 expression is related to increased response rate and clinical benefit in anti-PD-1/anti-PD-L1 therapy [28, 29]. In the phase 2 study Keynote-052, patients with urothelial cancer were treated with pembrolizumab, and increased positive predictive value was obtained along with increased PD-L1 expression cutoff value in the range of 1–10% [30]. And the subgroup with PD-L1 expression above 10% showed higher objective response rate than subgroup with PD-L1 expression below 1% (39% vs. 11%) [30]. However, the correlation between elevated PD-L1 expression and higher response rate is overthrown in some trials. In the study Checkmate-032 which involved patients with urothelial cancer, no significant difference in objective response rate (24.0% vs. 26.2%) was observed between PD-L1 expression positive subgroup (≥1%) and negative subgroup (< 1%) [31].
Table 1
Clinical trials of PD-1/PD-L1 inhibitors
Agents
Tumors
PD-L1 IHC platforms
Cells scored by IHC
Cutoff
Efficacy of agent
Clinical trial
Ref.
Pembrolizumab
Urothelial cancer
Dako 22C3 pharmDx Assay
Combined score of TC and IC
< 1%:
11, 95%CI 4–24% (ORRa)
Keynote-052
[30]
1–9%
20, 95%CI 14–28%(ORRa)
≥10%
39, 95% CI 28–50% (ORRa)
Melanoma
Dako 22C3 pharmDx Assay
Combined score of TC and IC
< 1%:
36.4, 95% CI 10.9–69.2% (ORRb)
Keynote-041
[116]
≥1%
16.7%, 95% CI 3.6–41.4% (ORRb)
HNSCC
Dako 22C3 pharmDx Assay
Combined score of TC and IC
< 50%:
13, 95%CI 7–20% (ORRb)
Keynote-055
[117]
≥50%
27, 95%CI 15–42% (ORRb)
NSCLC
Dako 22C3 pharmDx Assay
TC
< 1%
8.3, 95% CI 0.2–38.5%(ORRa)
Keynote-001
[118]
1–49%
17.3, 95% CI 8.2–30.3% (ORRa)
≥50%
51.9, 95% CI 31.9–71.3% (ORRa)
Melanoma
Dako 22C3 pharmDx Assay
Combined score of TC and IC
< 1%
2.8moths, 95% CI 2.7–2.8 months (PFS) 12.6 months, 95% CI 7.0–18.5 months (OS)
Keynote-001
[119]
≥1%
5.6 months, 95% CI 4.4–8.1 months (PFS) 29.9 months, 95% CI 24.6-NR months (OS)
Nivolumab
Squamous NSCLC
Dako 28–8 pharmDx Assay
TC
< 1%
HR of 2 years OS between Nivolumab and Docetaxel
HR:0.57, 95%CI 0.38–0.86
Checkmate-017
[120]
≥1%
HR:0.75, 95%CI 0.50–1.10
≥5%
HR:0.57, 95%CI 0.36–0.92
≥10%
HR:0.56, 95%CI 0.33–0.94
≥50%
HR:0.63, 95%CI 0.25–1.57
Non-squamous NSCLC
Dako 28–8 pharmDx Assay
TC
< 1%
HR of 2 years OS between Nivolumab and Docetaxel
HR:0.91, 95%CI 0.67–1.22
Checkmate-057
[120]
≥1%
HR:0.62, 95%CI 0.47–0.83
≥5%
HR:0.48, 95%CI 0.34–0.68
≥10%
HR:0.43, 95%CI 0.30–0.62
≥50%
HR:0.38, 95%CI 0.24–0.60
Urothelial cancer
Dako 28–8 pharmDx Assay
TC
< 1%
16.1, 95% CI 10.5–23.1% (ORRa)
Checkmate-275
[121]
≥1%
23.8, 95% CI 16.5–32.3% (ORRa)
≥5%
28.4, 95% CI 18.9–39.5% (ORRa)
Urothelial cancer
Dako 28–8 pharmDx Assay
TC
< 1%
26.2%; 95% CI 13.9–42.0%(ORRa)9·9 months, 95% CI 7.0–not estimable (median OS)
Checkmate-032
[31]
≥1%
24.0%; 95% CI 9.4–45.1% (ORRa)16.2 months, 95% CI 7.6–NE (median OS)
Renal cell cancer
Dako Assayc
TC
< 1%
HR of median OS between Nivolumab and everolimusHR: 0.76; 95% CI 0.60–0.97
Checkmate-025
[122]
≥1%
HR: 0.78; 95% CI 0.53–1.16
Squamous NSCLC
Dako Assayc
TC
< 5%
Best overall response 14% (PR),20% (SD),49% (PD)
Checkmate-063
[123]
≥5%
Best overall response 24% (PR),24% (SD),44% (PD)
Renal cell cancer
Dako 28–8 pharmDx Assay
TC
< 5%
18% (ORRa)2.9 months (median PFS)
NCT01354431.
[124]
≥5%
31% (ORRa)4.9 months (median PFS)
Melanoma
Dako Assayc
TC
< 5% or undefined
ORRa: 33.1, 95% CI 25.2–41.7% vs. 15.7%,
95% CI 10.0–23.0% (nivolumab vs. dacarbazine)
Checkmate-066
[125]
≥5%
ORRa: 52.7, 95% CI 40.8–64.3% vs. 10.8%,
95% CI 4.8–20.2% (nivolumab vs. dacarbazine)
Multiple cancers
IHC staining with anti-PD-L1 mAb 5H1
TC
< 5%
0% (ORRa)
NCT00730639
[126]
≥5%
36% (ORRa)
Atezolizumab
NSCLC
Ventana SP142 assay
TC or IC
TC and IC < 1%
HR of OS between atezolizumab and docetaxel HR:0.75, 95% CI 0.59–0.96
OAK
[127]
TC or IC ≥ 1%
HR:0.74, 95% CI 0.58–0.93
Urothelial cancer
Ventana SP142 assay
IC
< 1%
21, 95%CI 9–37% (ORRa)
NCT02108652
[128]
1–4%
21, 95%CI 11–35%(ORRa)
≥5%
28, 95%CI 14–47% (ORRa)
Renal cell cancer
Ventana SP142 assay
IC
< 1%
9, 95%CI 1–29% (ORRa)51, 95%CI 27–74% (2-Years OS Rate)
NCT01375842
[129]
≥1%
18, 95%CI7–35% (ORRa)65, 95%CI45–86% (2-Years OS Rate)
Multiple cancers
Ventana SP142 assay
IC
< 1%
13%(ORRa), 24-weeks PFS:33.9%
NCT01375842
[130]
1–4%
21% (ORRa), 24-weeks PFS:40.9%
5–9%
17% (ORRa), 24-weeks PFS:43.0%
≥10%
46% (ORRa), 24-weeks PFS:60.0%
NSCLC
Ventana SP142 assay
TC or IC
TC and IC < 1%
HR of OS between atezolizumab and docetaxel:1.04, 95%CI 0.62–1.75
POPLAR
[131]
TC or IC ≥1%
HR of OS between atezolizumab and docetaxel: 0.59, 95%CI 0.40–0.85
TC or IC ≥5%
HR of OS between atezolizumab and docetaxel: 0.54, 95%CI 0.33–0.89
TC ≥50% or IC ≥10%
HR of OS between atezolizumab and docetaxel: 0.49, 95%CI 0.22–1.07
Durvalumab
Urothelial cancer
Ventana SP263 assay
TC or IC
TC and IC < 25%
5.1, 95%CI 1.4–12.5%(ORRa)
NCT01693562
[132]
TC or IC ≥25%
27.6,95%CI 19.0–37.5%(ORRa)
Avelumab
Urothelial cancer
Dako assay
TC
< 5%
4.2% (ORRb)12 months-OS rate: 56.3, 95%CI 33.7–73.9%
NCT01772004
[133]
≥5%
53.8% (ORRb)12 months-OS rate: 75.5, 95%CI 41.6–91.4%
Abbreviations: CI confidence interval, HNSCC head and neck squamous cell carcinoma, HR hazard ratio, IC tumor infiltrating immune cell, NE not estimable, ORRa objective response rate, ORRb overall response rate, OS overall survival, mAb monoclonal antibody, PD, progressive disease, PFS progressive-free-survival, PR partially response, SD stably disease, TC tumor cell, Dako Assayc anti-PD-L1 antibody is not given
Many hypotheses have been put forward to explain the difference. Firstly, as the immunohistochemistry (IHC) is widely adopted in detection of PD-L1 expression, different cutoff values and scoring systems are used in separate clinical trials [24, 32]. And different antibodies and IHC platforms lead to the incomparability of results among trials as well [33]. Moreover, upregulated PD-L1 expression could be attributed to multiple causes. Intracellular oncogenic variations such as loss of PTEN and exposure to TIL-derived cytokines both contribute to upregulated PD-L1 expression [34]. However, immunity dependent PD-L1 upregulation is more meaningful to reactivate the tumor killing activity of TIL while intracellular oncogenic signaling pathway mediated upregulated PD-L1 has limited predictive value [34]. Lastly, due to intratumoral heterogeneity and dynamic alteration of PD-L1 expression along with treatment and cancer progression, the actual status of PD-L1 would be misinterpreted [35, 36].

The predictive value of PD-L1 expression in combination therapy

In spite of many limitations mentioned above, PD-L1 status is still a core predictor of treatment effect. However, this viewpoint is challenged in the context of combination strategy. A recent clinical trial interrogated the efficacy of combination strategy including atezolizumab, bevacizumab, carboplatin, and paclitaxel (ABCP) in metastatic non-squamous NSCLC patients [37]. Prognosis of patients receiving ABCP was improved significantly compared with treatment consisting of bevacizumab, carboplatin, and paclitaxel (BCP) [37]. Notably, for patients without epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) variations, ABCP group had prolonged RFS (HR = 0.77, p < 0.05, in PD-L1 patients) and OS (HR = 0.78, p = 0.02, in PD-L1 and PD-L1+ patients) regardless of PD-L1 status in comparison with BCP group [37]. Due to enhanced migration of neoantigen specific T cell and attenuated immune suppression caused by anti-angiogenesis and other treatments, it is difficult to predict alteration of immune microenvironment of PD-L1 patient post combination treatment [38, 39]. Therefore, in the context of combination of multiple drugs, the predictive value of PD-L1 expression is vague and deserves further investigation.

The heterogeneity of PD-L1 expression

Heterogeneous distribution of PD-L1+ tumor or stromal cell results in discordance between biopsy specimen and resection tissue [40]. Therefore, when resection tissue is not available, especially for some advanced cancer patients, PD-L1 expression of the whole tumor microenvironment might be displayed inaccurately [40, 41]. In the meanwhile, the probability of false negative event is increased. Notably, multiple cores biopsy showed higher sensitivity for selection of PD-L1+ patients compared with single core biopsy [40]. Besides, expression of PD-L1 variates during cancer evolution and treatment which is another obstacle to profiling immune microenvironment landscape. Kelly RJ et al. found that a significant shift from PD-L1 to PD-L1+ status in 50% advanced esophageal adenocarcinoma patients post chemo-radiation (OR = 6.5, p < 0.01) [42]. Tumor immune environment is subject to the influence of multiple factors, which determines the balance of immune surveillance and tolerance status.

TIL

TIL is a vital component influencing tumor immune microenvironment. Furthermore, TIL density has been confirmed to associate with adaptive upregulation of PD-L1 and clinical benefits [43]. Pre-existing TIL is unleashed by PD-1/PD-L1 inhibitors and then contributes to tumor regression [44, 45]. Recently, a tumor immune microenvironment model which consists of TIL status (presence or absence) and PD-L1 expression status (positive or negative) is established for immunotherapy prediction [46]. Cancer patients are classified into four types in the model, and Type I (PD-L1+TIL+) tumor is most likely to respond to PD-1/PD-L1 blockade therapy [46]. However, Type III (PD-L1+TIL) tumor is prone to resist to monotherapy of PD-1/PD-L1 inhibitors while the combination of PD-1/PD-L1 inhibitors and adjuvant therapy recruiting T cell into tumor bed would help to reverse the resistance [46]. CD8+ TIL is believed to be a vital player in killing tumor cell directly and maintaining the immune surveillance which could be spoilt by the signaling produced by PD-1/PD-L1 axis [47]. Solomon B et al. found that high density of CD8+ TIL was related with prolonged OS (HR: 0.4, 95%CI: 0.2–0.9, p = 0.017) [47].
Simultaneously, in another model based on the status of TIL, tumor immune microenvironment is classified into three subtypes: immune inflamed subtype, excluded infiltrate subtype, and immune ignorance subtype [48]. Recently, transforming growth factor β (TGF-β) signaling pathway attracts extensive attention because of its influence on T cell infiltration and distribution in tumor bed [4951]. Mariathasan S et al. conducted a study which enrolled metastatic urothelial cancer patients receiving atezolizumab treatment [49]. In the study, it was noticed that infiltration of T cell into tumor bed might be hampered by activated TGF-β signaling pathway in peritumoral fibroblast (Fig. 1) [49]. And simultaneously, tumor-specific T cell tended to distribute in peritumoral stroma rather than in intratumoral parenchyma [49]. The combined application of TGF-β signaling pathway blockade and PD-1/PD-L1 blockade had the significant advantage in tumor control with conversion of tumor environment from excluded infiltrate subtype to immune inflamed subtype [4951]. Notably, high pan fibroblast TGF-β response signature (TGF-β, TGF-β receptor, etc.) is related with non-response and tumor progression, especially for patient belonging to excluded infiltrate subtype [49].

TIL derived interferon-γ (IFN-γ)

IFN-γ signaling pathway is a double-edged sword in immune surveillance. On the one hand, CD8+ T cell inhibits tumor cell proliferation and enhances immune activity by secreting IFN-γ. On the other hand, T cell-derived IFN-γ upregulates PD-L1 expression on tumor cell as a shield to protect tumor cells from the immune surveillance’s attack [52, 53]. Upregulated PD-L1 driven by IFN-γ is the hallmark of potential tumor killing activation which is corresponded to Type I (PD-L1+TIL+) tumor above-mentioned. IFN-γ expression is generally believed to predict a favorable immune microenvironment to anti-PD-1/PD-L1 therapy [54]. IFNG mRNA expression extracted from formalin-fixed paraffin-embedded tissue specimens is positively related with the effect of anti-PD-1/PD-L1 treatment [55]. However, with PD-1/PD-L1 blockade, constant exposure to IFN-γ leads to survival selective pressure that tumor cells with defect in IFN-γ signaling pathway are most likely to proliferate (Fig. 2) [56]. Loss of downstream signals of IFN-γ is related to adaptive drug resistance during immunotherapy [52]. As a consequence, intact IFN-γ signaling pathway is a necessary but non-sufficient determinant for robust anti-tumor effect.
In fact, apart from IFN-γ, other inflammatory cytokines could induce adaptive immune resistance in multiple cancers. Tumor necrosis factor-α (TNF-α) mediates the de-differentiation of melanoma cell [13]. Moreover, TNF-α, Interleukin-6 (IL-6), and TGF-β are related to epithelial-to-mesenchymal transition (EMT) in multiple cancers such as melanoma and breast cancer [57, 58]. Notably, the cross-talk between TGFβ/TGFβRII pathway and PD-1/PD-L1 axis has been verified to contribute to T cell anergy in transplantation tolerance, but the mechanism should be investigated in tumor immune microenvironment further [59].

Tumor intrinsic feature related biomarkers

Tumor mutational burden

As a biomarker independent of PD-L1 expression, accumulated mutations with increased potentiality of neoantigen results in elevated immunogenicity (Fig. 3) [60, 61]. Correspondingly, activated immune microenvironment is favorable to tumor shrink in the context of anti-PD-1/PD-L1 treatment [62]. Based on Next-Generation Sequencing, it is available to profile nonsynonymous somatic mutations of tumor cell [63]. The level of tumor mutational burden (TMB) is evaluated by mutations per megabase [60]. A pooled analysis involving 27 tumor types/subtypes revealed a significant correlation between TMB and objective response rate (correlation coefficient: 0.74) [64]. Notably, clonal mutations (shared by all tumor cells) and subclonal mutations (expressing on a fraction of tumor cells) affect tumor specific immunity differently [65]. McGranahan N et al. found that homogeneous tumor with high TMB associated with increased clinical benefits and sensitivity to anti-PD-1/PD-L1 therapy [65]. However, tumor with high subclonal mutation rate tends to accompany poor anti-PD-1/PD-L1 effect [60]. Single-site biopsy might overestimate level of clonal mutation due to the interference from subclonal mutation which might explain the poor response of some patients with high TMB [62, 65].

Mismatch repair deficiency and microsatellite instability

Mismatch repair (MMR) system participates in rectifying base-base mismatch, insertion, and deletion defect during DNA replication [66]. Members belonging to MMR system including MutL homolog 1 (MLH1), MutS protein homolog 2 (MSH2), MutS homolog 6 (MSH6), and PMS1 homolog 2 (PMS2) contribute to maintaining genomic stability while reduction or depletion of MMR promotes oncogenesis, especially in gastrointestinal cancers [63, 67]. Mismatch repair deficiency (dMMR) leads to the accumulation of mutation as well as production of potential neoantigen (Fig. 3). Furthermore, MMR IHC and microsatellite instability (MSI) analyzed by Polymerase Chain Reaction (PCR) revealed a high concordance between dMMR and MSI [68]. In fact, the primary reason of MSI is epigenetic or genetic variation of MMR [69, 70]. Xiao X et al. found existence of MSI in all ovarian cancer patients with dMMR [71]. MSI-high (MSI-H)/dMMR associates with favorable prognosis of patients receiving anti-PD-1/PD-L1 therapy [72]. Kumar R et al. observed that anti-PD-1 promoted dMMR tumor cell apoptosis by cytotoxicity of CD8+ T cell in vitro in comparison with MMR proficient tumor cell [73]. Le DT et al. conducted a study to explore the influence of MSI-H on anti-PD-L1 therapy, and satisfactory treatment effect was observed (objective radiographic response rate: 53%, complete response rate: 21%) in multiple cancer patients with dMMR [74]. Enhanced treatment effect resulting from MSI-H/dMMR is attributed to increased density of TIL, elevated TMB, upregulated PD-L1 expression, and more potent tumor-specific immune response [72, 75, 76].

Oncogenic driver mutations and other mutations

It has been found that some driver mutations affect PD-L1 expression such as mutation of EGFR, Kirsten rat sarcoma viral oncogene homology (KRAS), and ALK [77]. EGFR activating mutation (mEGFR) upregulates PD-L1 expression and impedes the activation of TIL [78]. Contrary to expectation, patients harboring mEGFR tends to have poorer response in comparison with patients with wild EGFR during anti-PD-1/PD-L1 therapy. PD-L1 expression could be regulated by both extracellular immune factor and intracellular oncogenic driver signal. Given the activated EGFR-mediated PD-L1 expression by PI3K-AKT-STAT3/mTOR signaling pathways as well as simultaneous mEGFR-induced IFN-γ decline, it is hard to estimate whether PD-L1 expression is regulated just depending on EGFR status [78, 79]. Besides, mEGFR is relevant to low TMB and compromised tumor-specific immune response [78]. In contrast to mEGFR, meta-analysis revealed that NSCLC patients harboring KRAS mutation are more likely to belong to PD-L1 positive subtype [80]. And Coelho MA et al. found that hyperactive KRAS enhanced stability of PD-L1 mRNA by MEK-ERK signal pathway [81]. Notably, co-occurring mutation with mutated KRAS affects tumor microenvironment in different ways. Mutated KRAS with co-occurring serine/threonine kinase 11/liver kinase B1 variation associates with upregulated expression of PD-L1 while co-occurring mutation with TP53 accompanies high TMB abundance [81]. Moreover, ALK arrangement in inflammatory myofibroblastic tumor is related to decreased CD8+ TIL as well as downregulated PD-L1 expression [82]. Except for driver mutations, some other somatic mutations modulate tumor-specific immune response as well. Kataegis is a special mutation pattern which is caused by variation of apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) [83]. Boichard A et al. found that Kataegis and APOBEC3 overexpression participated in regulation of PD-L1 expression [83]. Furthermore, polymerase δ1 (POLD1) and polymerase ε (POLE) variations lead to extremely high frequency of somatic mutation which affects tumor immunogenicity [84, 85].

Gut microbiota

Cross-talk between gut microbiota and host immunity influences anti-tumor effect of anti-PD-1/PD-L1 therapy and the predictive value of gut microbiota has been noticed recently (Table 2) [86]. Using mouse xenograft model, Ayelet Sivan et al. observed that fecal microbiome transplantation could restore the sensitivity to anti-PD-L1 treatment and improve anti-tumor activity in non-responding mice [87]. And increased Bifidobacterium abundance accounts for the alteration mentioned above [87]. Besides, Gopalakrishnan V et al. noticed the relationship between high abundance of Faecalibacterium genus and elevated response rate in patients receiving anti-PD-1 treatment [88]. In the meanwhile, dysbacteriosis caused by utilization of antibiotics was proved to influence the efficacy of anti-PD-1/PD-L1 therapy. And the poor response to agents could be reversed by recolonization of Akkermansia muciniphila [89]. Though the exact modulatory mechanism is unclear, many factors are proposed to enhanced tumor control (Fig. 3). Firstly, Bifidobacterium promotes maturation and activation of dendritic cell (DC) which enhances neoantigen presentation process [87]. Secondly, recolonization of Akkermansia muciniphila and Enterococcus hirae associates with appearance of CD4+ central memory T cell (TCM) in tumor bed [89]. And TCM leads to increased CD4/Foxp3+ ratio in tumor bed by enhancing recruitment and chemotactic migration of T cell [89]. Thirdly, bacteria could be sensed by host immunity and then influences the differentiation of lymphocytes such as Th1 and pTh17 in second immune organ. The alteration of microbiota composition might change the tumor immune microenvironment by the homing and recirculation of lymphocytes [89, 90]. Furthermore, bacterial metabolites such as short chain fatty acid (SCFA) participates in energy metabolism of immune cell which might affects the function of immunity [91]. Finally, potential molecular mimicry between gut microbiota and tumor might participates in tumor-specific immune response [92]. Therefore, analyzing gut microbiota composition would be favorable to predict treatment effect of anti-PD-1/PD-L1 therapy.
Table 2
The role of gut microbiota in PD-1/PD-L1 inhibitors therapy
Bacteria
Main effect on immunity
Prediction of treatment effect
Model
Ref.
A. muciniphila
Increased recruitment of CCR9+CXCR3+CD4+ T cells into tumor bed
Effective anti-tumor response
Mouse/Human
[89]
E. hirae
Increased IL-12 secreted by DC
Effective anti-tumor response
Mouse/Human
[89]
E. faeciumC. aerofaciens
B. adolescentisK. pneumoniae
V. parvulaP. merdae
Lactobacillus sp. B. longum
Increased neoantigen specific CD8+ T cell and decreased Fox3P+CD4+ Treg in tumor microenvironment
Effective anti-tumor response
Mouse/Human
[134]
Bifidobacterium
Increased IFN-γ production and major histocompatibility complex Class IIhi DC
Effective anti-tumor response
Mouse
[87]
Faecalibacterium
Increased peripheral effector CD4+ and CD8+ T cell
Effective anti-tumor response
Mouse/Human
[88]
Bacteroidales
Increased peripheral Treg and myeloid derived suppressor cell
Poor anti-tumor response
Mouse/Human
[88]
Abbreviations: IFN-γ interferon-γ, Treg regulatory T cell, DC dendritic cell

Biomarkers in peripheral blood

Compared with biopsy sample from tumor tissue, peripheral blood sample is more available and less heterogeneous. Due to negligible invasion, it is an ideal access to monitor shift of biomarkers in peripheral blood for optimized therapy strategy (Fig. 3) [93].

Peripheral immune cell

Using mass cytometry and bioinformatics analysis, Krieg C et al. observed that high abundance of peripheral CD14+CD16HLA-DRhi monocyte at baseline associated with higher response rate in anti-PD-1/PD-L1 therapy. And the increased markers on membrane such as intercellular cell adhesion molecule-1 (ICAM-1) and human leukocyte antigen-antigen D related (HLA-DR) indicate enhanced migration and activation of monocyte. Besides, responding patients tended to have decreased T cell in peripheral blood in comparison with non-responding patients. Supposedly, CD14+CD16HLA-DRhi monocyte promotes the infiltration of T cell from peripheral blood into tumor bed which results in enhanced T cell-mediated tumor killing activity [94]. Besides, Kamphorst AO et al. noticed that early expansion of peripheral PD-1+Ki-67+CD8+ T cells after anti-PD-1 treatment was related to better treatment effect. And peripheral PD-1+Ki-67+CD8+ T cell was detected to express more activation-associated markers such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and inducible T cell costimulator (ICOS) [95]. Furthermore, Fujisawa Y et al. found that neutrophil/lymphocyte ratio and lactate dehydrogenase (LDH) level associated with response to nivolumab in melanoma patients. Elevated neutrophil/lymphocyte ratio (> 2.2) predicted poor treatment effect (OR = 4.16, p = 0.0026) while increased peripheral LDH was related with poor response tendency without statistical significance (OR = 2.53, p = 0.081) [96]. Contrary to neutrophil, increased relative eosinophil count (≥1.5%) could be used as a favorable predictor in melanoma patients receiving pembrolizumab [97].

Circulating tumor DNA and PD-L1high circulating tumor cell

Radiological assessment is widely applied to evaluate the treatment effect of anti-PD-1/PD-L1. However, interference of pseudo-progression and non-real time reflection of tumor burden might affect the selection of subsequent treatment strategy [98]. It was observed that circulating tumor DNA (ctDNA) in responding patient decreased quickly in 5 days after first nivolumab administration. The phenomenon is meaningful that the shift in ctDNA is prior to second administration and radiological change [99]. Compared with detectable abundance at baseline, undetectable ctDNA after therapy beginning indicates robust anti-tumor effect which is valuable for early patient selection [98, 100]. Similarly, decreased PD-L1+ circulating tumor cell after treatment beginning is related to robust anti-tumor response. However, patients with high abundance of PD-L1+ circulating tumor cell at baseline tend to be sensitive to anti-PD-L1 therapy [101].

Soluble PD-L1

Splice variants of PD-L1 which lack transmembrane or intracellular domain lead to secretion of soluble PD-L1 (sPD-L1) [102]. Similar to membrane-binding PD-L1, sPD-L1 hampers the activation and proliferation of T cell as well [103]. It is generally acknowledged that increased level of sPD-L1 before treatment associates with poor prognosis which is attributed to high tumor burden, elevated alternative splicing, and exhausted immune response [102, 104]. Zhou J et al. found that high sPD-L1 at baseline was related with increased risk of tumor progression. However, rapidly increased sPD-L1 level after immune checkpoint inhibitors treatment indicated potent tumor-specific immune response and high partial response rate (around 70%) [102].

Peripheral cytokine and other parameters

Peripheral cytokines reflect status of tumor immune microenvironment and response to anti-PD-1/PD-L1 treatment [93]. Prolactin (PRL) participates in maturation and activation of immunity while high PRL inhibits immune response by IL-10 [105]. Adaptive hyperprolactinemia associates with poor response during nivolumab treatment and patients with stable concentration of PRL exhibit significant higher response rate (p = 0.004) [105]. Moreover, a phase 2 study revealed that pretreatment high level of IFN-γ, IL-6, and IL-10 in peripheral blood were relevant to increased objective response rate in melanoma patients receiving nivolumab [106]. Besides, tumor-derived vascular endothelial growth factor (VEGF) promotes tumor progression by angiogenesis and immunosuppression in tumor microenvironment [107]. Anti-angiogenesis therapy not only inhibits neo-vascular formation, but also upregulates the quantity of TIL significantly [108]. Patients receiving anti-PD-L1 combined with anti-VEGF therapy exhibited higher response rate than monotherapy [107, 109]. Cytokines participate in immune response directly, and the predictive value of cytokine in peripheral blood needs to explore further.

Patient previous history, pathological feature, and other predictors

Chronic obstructive pulmonary disease (COPD) participates in oncogenesis and COPD-associated chronic inflammation influences immune environment of lung cancer patient in the meanwhile [34]. Biton J et al. interrogated treatment response of lung cancer patients receiving nivolumab. Lung cancer patients with co-existing COPD tended to harbor higher inhibitory markers such as PD-1 and TIM-3, which indicated more severe exhaustion of TIL in comparison with patients without COPD [34, 110]. NSCLC patients with co-existing COPD had favorable prognosis during nivolumab treatment and increased correlation between PD-L1 expression and response rate [34]. Notably, cigarette exposure contributes to oncogenesis of lung cancer as well as occurrence of COPD [111]. Because cigarette exposure leads to increased TMB which might cause enhanced the sensitivity to immunotherapy, it is necessary to rule out the interference from cigarette exposure [112]. By analyzing TMB, KRAS, and TP53 variations in COPD+ patients, no significant enrichment of smoking signature was observed in COPD+ patients [34]. Therefore, COPD is speculated as a potential predictor for anti-PD-1/PD-L1 treatment. Besides, immune microenvironment alters among tumors with different pathological features. In three subtypes of lung adenocarcinoma, the level of TMB and immune cell signature change significantly [113]. Tumor belonging to proximal inflammatory subtype tends to have higher TMB, TP53 variation, and immune cell signature, while tumor belonging to terminal respiratory unit subtype is most likely to harbor low TMB without TP53 mutation [113]. And the predictive value of pathological feature needs to be verified in large sample size. Intriguingly, a recent pilot study revealed the correlation between family history of cancer and treatment effect of anti-PD-1/PD-L1 therapy [114]. Multiple cancers patients with family history of cancer had significantly improved objective response rate (p = 0.0024) and favorable outcome [114].

Conclusion

PD-L1 expression is generally believed as a surrogate of pre-existing immune specific immune activity and can be upregulated by IFN-γ in tumor microenvironment [115]. However, other factors simultaneously influence PD-L1 expression such as intracellular oncogenic signaling pathway apart from adaptive immune resistance. Therefore, total PD-L1 including IFN-γ-derived and IFN-γ-independent PD-L1 is not accurate to reflect tumor immune surveillance status [115]. Combination of PD-L1 expression, TIL, TMB, genetic and epigenetic variation of IFN-γ provides a comprehensive prospective on tumor immune landscape. Moreover, circulating biomarkers and gut microbiota play a vital role in dynamic monitoring of tumor immune status due to minimum invasion. With the increased understanding of tumor immune escape, establishing a wide-ranging framework which consists of multiple biomarkers is quite necessary for patient selection and precision medicine.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81572608, 81172422), Wuhan Science and Technology Bureau (No. 2017060201010170), and the National High Technology Research and Development Program of China (No. 2015AA020301).
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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.
Literatur
1.
2.
3.
Zurück zum Zitat Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, et al. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp Hematol Oncol. 2017;6:31.PubMedPubMedCentralCrossRef Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, et al. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp Hematol Oncol. 2017;6:31.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Crist M, Balar A. Atezolizumab in invasive and metastatic urothelial carcinoma. Expert Rev Clin Pharmacol. 2017;10:1295–301.PubMedCrossRef Crist M, Balar A. Atezolizumab in invasive and metastatic urothelial carcinoma. Expert Rev Clin Pharmacol. 2017;10:1295–301.PubMedCrossRef
5.
Zurück zum Zitat Kythreotou A, Siddique A, Mauri FA, Bower M, Pinato DJ. PD-L1. J Clin Pathol. 2018;71:189–94.PubMedCrossRef Kythreotou A, Siddique A, Mauri FA, Bower M, Pinato DJ. PD-L1. J Clin Pathol. 2018;71:189–94.PubMedCrossRef
6.
7.
Zurück zum Zitat Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5 ra46 Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5 ra46
10.
Zurück zum Zitat Kuol N, Stojanovska L, Nurgali K, Apostolopoulos V. PD-1/PD-L1 in disease. Immunotherapy. 2018;10:149–60.PubMedCrossRef Kuol N, Stojanovska L, Nurgali K, Apostolopoulos V. PD-1/PD-L1 in disease. Immunotherapy. 2018;10:149–60.PubMedCrossRef
11.
Zurück zum Zitat Juchem KW, Sacirbegovic F, Zhang C, Sharpe AH, Russell K, McNiff JM, et al. PD-L1 prevents the development of autoimmune heart disease in graft-versus-host disease. J Immunol. 2018;200:834–46.PubMedCrossRef Juchem KW, Sacirbegovic F, Zhang C, Sharpe AH, Russell K, McNiff JM, et al. PD-L1 prevents the development of autoimmune heart disease in graft-versus-host disease. J Immunol. 2018;200:834–46.PubMedCrossRef
12.
Zurück zum Zitat Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature. 2018;553:91–5.PubMedCrossRef Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature. 2018;553:91–5.PubMedCrossRef
14.
Zurück zum Zitat Witt DA, Donson AM, Amani V, Moreira DC, Sanford B, Hoffman LM, et al. Specific expression of PD-L1 in RELA-fusion supratentorial ependymoma: implications for PD-1-targeted therapy. Pediatr Blood Cancer. 2018;65:e26960.PubMedCrossRef Witt DA, Donson AM, Amani V, Moreira DC, Sanford B, Hoffman LM, et al. Specific expression of PD-L1 in RELA-fusion supratentorial ependymoma: implications for PD-1-targeted therapy. Pediatr Blood Cancer. 2018;65:e26960.PubMedCrossRef
15.
Zurück zum Zitat Zheng B, Ren T, Huang Y, Sun K, Wang S, Bao X, et al. PD-1 axis expression in musculoskeletal tumors and antitumor effect of nivolumab in osteosarcoma model of humanized mouse. J Hematol Oncol. 2018;11:16.PubMedPubMedCentralCrossRef Zheng B, Ren T, Huang Y, Sun K, Wang S, Bao X, et al. PD-1 axis expression in musculoskeletal tumors and antitumor effect of nivolumab in osteosarcoma model of humanized mouse. J Hematol Oncol. 2018;11:16.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Xu-Monette ZY, Zhang M, Li J, Young KH. PD-1/PD-L1 blockade: have we found the key to unleash the antitumor immune response? Front Immunol. 2017;8:1597.PubMedPubMedCentralCrossRef Xu-Monette ZY, Zhang M, Li J, Young KH. PD-1/PD-L1 blockade: have we found the key to unleash the antitumor immune response? Front Immunol. 2017;8:1597.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Karlsson AK, Saleh SN. Checkpoint inhibitors for malignant melanoma: a systematic review and meta-analysis. Clin Cosmet Investig Dermatol. 2017;10:325–39.PubMedPubMedCentralCrossRef Karlsson AK, Saleh SN. Checkpoint inhibitors for malignant melanoma: a systematic review and meta-analysis. Clin Cosmet Investig Dermatol. 2017;10:325–39.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Liu B, Song Y, Liu D. Recent development in clinical applications of PD-1 and PD-L1 antibodies for cancer immunotherapy. J Hematol Oncol. 2017;10:174.PubMedPubMedCentralCrossRef Liu B, Song Y, Liu D. Recent development in clinical applications of PD-1 and PD-L1 antibodies for cancer immunotherapy. J Hematol Oncol. 2017;10:174.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for Cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.PubMedPubMedCentralCrossRef Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for Cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Polk A, Svane IM, Andersson M, Nielsen D. Checkpoint inhibitors in breast cancer - current status. Cancer Treat Rev. 2017;63:122–34.PubMedCrossRef Polk A, Svane IM, Andersson M, Nielsen D. Checkpoint inhibitors in breast cancer - current status. Cancer Treat Rev. 2017;63:122–34.PubMedCrossRef
23.
Zurück zum Zitat Sheng Z, Zhu X, Sun Y, Zhang Y. The efficacy of anti-PD-1/PD-L1 therapy and its comparison with EGFR-TKIs for advanced non-small-cell lung cancer. Oncotarget. 2017;8:57826–35.PubMedPubMedCentral Sheng Z, Zhu X, Sun Y, Zhang Y. The efficacy of anti-PD-1/PD-L1 therapy and its comparison with EGFR-TKIs for advanced non-small-cell lung cancer. Oncotarget. 2017;8:57826–35.PubMedPubMedCentral
24.
Zurück zum Zitat Ancevski Hunter K, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung Cancer. Mol Diagn Ther. 2018;22:1–10.PubMedCrossRef Ancevski Hunter K, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung Cancer. Mol Diagn Ther. 2018;22:1–10.PubMedCrossRef
25.
Zurück zum Zitat Janjigian YY, Sanchez-Vega F, Jonsson P, Chatila WK, Hechtman JF, Ku GY, et al. Genetic predictors of response to systemic therapy in Esophagogastric Cancer. Cancer Discov. 2018;8:49–58.PubMedCrossRef Janjigian YY, Sanchez-Vega F, Jonsson P, Chatila WK, Hechtman JF, Ku GY, et al. Genetic predictors of response to systemic therapy in Esophagogastric Cancer. Cancer Discov. 2018;8:49–58.PubMedCrossRef
26.
Zurück zum Zitat Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–71.PubMedPubMedCentralCrossRef Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–71.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 2018;11:31.PubMedCrossRef Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 2018;11:31.PubMedCrossRef
28.
Zurück zum Zitat Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung Cancer. N Engl J Med. 2016;375:1823–33.PubMedCrossRef Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung Cancer. N Engl J Med. 2016;375:1823–33.PubMedCrossRef
29.
Zurück zum Zitat Ding W, LaPlant BR, Call TG, Parikh SA, Leis JF, He R, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood. 2017;129:3419–27.PubMedPubMedCentralCrossRef Ding W, LaPlant BR, Call TG, Parikh SA, Leis JF, He R, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood. 2017;129:3419–27.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18:1483–92.PubMedCrossRef Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18:1483–92.PubMedCrossRef
31.
Zurück zum Zitat Sharma P, Callahan MK, Bono P, Kim J, Spiliopoulou P, Calvo E, et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 2016;17:1590–8.PubMedPubMedCentralCrossRef Sharma P, Callahan MK, Bono P, Kim J, Spiliopoulou P, Calvo E, et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 2016;17:1590–8.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al. PD-L1 immunohistochemistry assays for lung Cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. 2017;12:208–22.PubMedCrossRef Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al. PD-L1 immunohistochemistry assays for lung Cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. 2017;12:208–22.PubMedCrossRef
35.
Zurück zum Zitat Li D, Chen R, Wang YW, Fornace AJ Jr, Li HH. Prior irradiation results in elevated programmed cell death protein 1 (PD-1) in T cells. Int J Radiat Biol. 2017;94:488–94.PubMedCrossRef Li D, Chen R, Wang YW, Fornace AJ Jr, Li HH. Prior irradiation results in elevated programmed cell death protein 1 (PD-1) in T cells. Int J Radiat Biol. 2017;94:488–94.PubMedCrossRef
36.
Zurück zum Zitat Vilain RE, Menzies AM, Wilmott JS, Kakavand H, Madore J, Guminski A, et al. Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin Cancer Res. 2017;23:5024–33.PubMedCrossRef Vilain RE, Menzies AM, Wilmott JS, Kakavand H, Madore J, Guminski A, et al. Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin Cancer Res. 2017;23:5024–33.PubMedCrossRef
37.
Zurück zum Zitat Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–301.PubMedCrossRef Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–301.PubMedCrossRef
38.
Zurück zum Zitat Bocca P, Di Carlo E, Caruana I, Emionite L, Cilli M, De Angelis B, et al. Bevacizumab-mediated tumor vasculature remodelling improves tumor infiltration and antitumor efficacy of GD2-CAR T cells in a human neuroblastoma preclinical model. Oncoimmunology. 2017;7:e1378843.PubMedCrossRefPubMedCentral Bocca P, Di Carlo E, Caruana I, Emionite L, Cilli M, De Angelis B, et al. Bevacizumab-mediated tumor vasculature remodelling improves tumor infiltration and antitumor efficacy of GD2-CAR T cells in a human neuroblastoma preclinical model. Oncoimmunology. 2017;7:e1378843.PubMedCrossRefPubMedCentral
39.
Zurück zum Zitat McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. 2018;24:749–57.PubMedCrossRef McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. 2018;24:749–57.PubMedCrossRef
43.
Zurück zum Zitat Xing X, Guo J, Wen X, Ding G, Li B, Dong B, et al. Analysis of PD1, PDL1, PDL2 expression and T cells infiltration in 1014 gastric cancer patients. Oncoimmunology. 2018;7:e1356144.PubMedCrossRef Xing X, Guo J, Wen X, Ding G, Li B, Dong B, et al. Analysis of PD1, PDL1, PDL2 expression and T cells infiltration in 1014 gastric cancer patients. Oncoimmunology. 2018;7:e1356144.PubMedCrossRef
45.
Zurück zum Zitat Tomioka N, Azuma M, Ikarashi M, Yamamoto M, Sato M, Watanabe KI, et al. The therapeutic candidate for immune checkpoint inhibitors elucidated by the status of tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression in triple negative breast cancer (TNBC). Breast Cancer. 2018;25:34–42.PubMedCrossRef Tomioka N, Azuma M, Ikarashi M, Yamamoto M, Sato M, Watanabe KI, et al. The therapeutic candidate for immune checkpoint inhibitors elucidated by the status of tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression in triple negative breast cancer (TNBC). Breast Cancer. 2018;25:34–42.PubMedCrossRef
48.
Zurück zum Zitat Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for Cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res. 2016;22:1865–74.PubMedCrossRef Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for Cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res. 2016;22:1865–74.PubMedCrossRef
49.
Zurück zum Zitat Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–8.PubMedPubMedCentralCrossRef Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–8.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFbeta enhance the efficacy of cancer immunotherapy. Nat Commun. 2018;9:741.PubMedPubMedCentralCrossRef Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFbeta enhance the efficacy of cancer immunotherapy. Nat Commun. 2018;9:741.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. 2018;11:39.PubMedPubMedCentralCrossRef Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. 2018;11:39.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Tremblay-LeMay R, Rastgoo N, Chang H. Modulating PD-L1 expression in multiple myeloma: an alternative strategy to target the PD-1/PD-L1 pathway. J Hematol Oncol. 2018;11:46.PubMedPubMedCentralCrossRef Tremblay-LeMay R, Rastgoo N, Chang H. Modulating PD-L1 expression in multiple myeloma: an alternative strategy to target the PD-1/PD-L1 pathway. J Hematol Oncol. 2018;11:46.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Teng F, Meng X, Kong L, Yu J. Progress and challenges of predictive biomarkers of anti PD-1/PD-L1 immunotherapy: a systematic review. Cancer Lett. 2018;414:166–73.PubMedCrossRef Teng F, Meng X, Kong L, Yu J. Progress and challenges of predictive biomarkers of anti PD-1/PD-L1 immunotherapy: a systematic review. Cancer Lett. 2018;414:166–73.PubMedCrossRef
55.
Zurück zum Zitat Karachaliou N, Gonzalez-Cao M, Crespo G, Drozdowskyj A, Aldeguer E, Gimenez-Capitan A, et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther Adv Med Oncol. 2018;10 1758834017749748 Karachaliou N, Gonzalez-Cao M, Crespo G, Drozdowskyj A, Aldeguer E, Gimenez-Capitan A, et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther Adv Med Oncol. 2018;10 1758834017749748
56.
Zurück zum Zitat Abril-Rodriguez G, Ribas A. SnapShot: Immune Checkpoint Inhibitors. Cancer Cell. 2017;31:848–e1.PubMedCrossRef Abril-Rodriguez G, Ribas A. SnapShot: Immune Checkpoint Inhibitors. Cancer Cell. 2017;31:848–e1.PubMedCrossRef
57.
Zurück zum Zitat Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature. 2012;490:412–6.PubMedCrossRef Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature. 2012;490:412–6.PubMedCrossRef
58.
Zurück zum Zitat Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, et al. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells. Cancer Res. 2009;69:2887–95.PubMedPubMedCentralCrossRef Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, et al. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells. Cancer Res. 2009;69:2887–95.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Baas M, Besancon A, Goncalves T, Valette F, Yagita H, Sawitzki B, et al. TGFbeta-dependent expression of PD-1 and PD-L1 controls CD8(+) T cell anergy in transplant tolerance. elife. 2016;5:e08133.PubMedPubMedCentralCrossRef Baas M, Besancon A, Goncalves T, Valette F, Yagita H, Sawitzki B, et al. TGFbeta-dependent expression of PD-1 and PD-L1 controls CD8(+) T cell anergy in transplant tolerance. elife. 2016;5:e08133.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Hodges TR, Ott M, Xiu J, Gatalica Z, Swensen J, Zhou S, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology. 2017;19:1047–57.PubMedPubMedCentralCrossRef Hodges TR, Ott M, Xiu J, Gatalica Z, Swensen J, Zhou S, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology. 2017;19:1047–57.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung Cancer profiled with targeted next-generation sequencing. J Clin Oncol. 2018;36:633–41.PubMedPubMedCentralCrossRef Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung Cancer profiled with targeted next-generation sequencing. J Clin Oncol. 2018;36:633–41.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–608.PubMedCrossRef Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–608.PubMedCrossRef
63.
Zurück zum Zitat Yuza K, Nagahashi M, Watanabe S, Takabe K, Wakai T. Hypermutation and microsatellite instability in gastrointestinal cancers. Oncotarget. 2017;8:112103–15.PubMedPubMedCentralCrossRef Yuza K, Nagahashi M, Watanabe S, Takabe K, Wakai T. Hypermutation and microsatellite instability in gastrointestinal cancers. Oncotarget. 2017;8:112103–15.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.PubMedCrossRef Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.PubMedCrossRef
65.
Zurück zum Zitat McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, 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, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.PubMedPubMedCentralCrossRef
66.
67.
Zurück zum Zitat Kim ST, Klempner SJ, Park SH, Park JO, Park YS, Lim HY, et al. Correlating programmed death ligand 1 (PD-L1) expression, mismatch repair deficiency, and outcomes across tumor types: implications for immunotherapy. Oncotarget. 2017;8:77415–23.PubMedPubMedCentral Kim ST, Klempner SJ, Park SH, Park JO, Park YS, Lim HY, et al. Correlating programmed death ligand 1 (PD-L1) expression, mismatch repair deficiency, and outcomes across tumor types: implications for immunotherapy. Oncotarget. 2017;8:77415–23.PubMedPubMedCentral
68.
Zurück zum Zitat Yamashita H, Nakayama K, Ishikawa M, Nakamura K, Ishibashi T, Sanuki K, et al. Microsatellite instability is a biomarker for immune checkpoint inhibitors in endometrial cancer. Oncotarget. 2018;9:5652–64.PubMed Yamashita H, Nakayama K, Ishikawa M, Nakamura K, Ishibashi T, Sanuki K, et al. Microsatellite instability is a biomarker for immune checkpoint inhibitors in endometrial cancer. Oncotarget. 2018;9:5652–64.PubMed
69.
Zurück zum Zitat Geiersbach KB, Samowitz WS. Microsatellite instability and colorectal cancer. Arch Pathol Lab Med. 2011;135:1269–77.PubMedCrossRef Geiersbach KB, Samowitz WS. Microsatellite instability and colorectal cancer. Arch Pathol Lab Med. 2011;135:1269–77.PubMedCrossRef
70.
Zurück zum Zitat Zeinalian M, Hashemzadeh-Chaleshtori M, Salehi R, Emami MH. Clinical aspects of microsatellite instability testing in colorectal Cancer. Adv Biomed Res. 2018;7:28.PubMedPubMedCentralCrossRef Zeinalian M, Hashemzadeh-Chaleshtori M, Salehi R, Emami MH. Clinical aspects of microsatellite instability testing in colorectal Cancer. Adv Biomed Res. 2018;7:28.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Xiao X, Dong D, He W, Song L, Wang Q, Yue J, et al. Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer. Gynecol Oncol. 2018;149:146–54.PubMedCrossRef Xiao X, Dong D, He W, Song L, Wang Q, Yue J, et al. Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer. Gynecol Oncol. 2018;149:146–54.PubMedCrossRef
72.
Zurück zum Zitat Jin Z, Yoon HH. The promise of PD-1 inhibitors in gastro-esophageal cancers: microsatellite instability vs. PD-L1. J Gastrointest Oncol. 2016;7:771–88.PubMedPubMedCentralCrossRef Jin Z, Yoon HH. The promise of PD-1 inhibitors in gastro-esophageal cancers: microsatellite instability vs. PD-L1. J Gastrointest Oncol. 2016;7:771–88.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Kumar R, Yu F, Zhen YH, Li B, Wang J, Yang Y, et al. PD-1 blockade restores impaired function of ex vivo expanded CD8(+) T cells and enhances apoptosis in mismatch repair deficient EpCAM(+)PD-L1(+) cancer cells. Onco Targets Ther. 2017;10:3453–65.PubMedPubMedCentralCrossRef Kumar R, Yu F, Zhen YH, Li B, Wang J, Yang Y, et al. PD-1 blockade restores impaired function of ex vivo expanded CD8(+) T cells and enhances apoptosis in mismatch repair deficient EpCAM(+)PD-L1(+) cancer cells. Onco Targets Ther. 2017;10:3453–65.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.PubMedPubMedCentralCrossRef Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Mills AM, Dill EA, Moskaluk CA, Dziegielewski J, Bullock TN, Dillon PM. The relationship between mismatch repair deficiency and PD-L1 expression in breast carcinoma. Am J Surg Pathol. 2018;42:183–91.PubMedCrossRef Mills AM, Dill EA, Moskaluk CA, Dziegielewski J, Bullock TN, Dillon PM. The relationship between mismatch repair deficiency and PD-L1 expression in breast carcinoma. Am J Surg Pathol. 2018;42:183–91.PubMedCrossRef
76.
Zurück zum Zitat El Jabbour T, Ross JS, Sheehan CE, Affolter KE, Geiersbach KB, Boguniewicz A, et al. PD-L1 protein expression in tumour cells and immune cells in mismatch repair protein-deficient and -proficient colorectal cancer: the foundation study using the SP142 antibody and whole section immunohistochemistry. J Clin Pathol. 2018;71:46–51.PubMedCrossRef El Jabbour T, Ross JS, Sheehan CE, Affolter KE, Geiersbach KB, Boguniewicz A, et al. PD-L1 protein expression in tumour cells and immune cells in mismatch repair protein-deficient and -proficient colorectal cancer: the foundation study using the SP142 antibody and whole section immunohistochemistry. J Clin Pathol. 2018;71:46–51.PubMedCrossRef
77.
Zurück zum Zitat Jiang L, Su X, Zhang T, Yin X, Zhang M, Fu H, et al. PD-L1 expression and its relationship with oncogenic drivers in non-small cell lung cancer (NSCLC). Oncotarget. 2017;8:26845–57.PubMedPubMedCentral Jiang L, Su X, Zhang T, Yin X, Zhang M, Fu H, et al. PD-L1 expression and its relationship with oncogenic drivers in non-small cell lung cancer (NSCLC). Oncotarget. 2017;8:26845–57.PubMedPubMedCentral
78.
Zurück zum Zitat Li X, Lian Z, Wang S, Xing L, Yu J. Interactions between EGFR and PD-1/PD-L1 pathway: implications for treatment of NSCLC. Cancer Lett. 2018;418:1–9.PubMedCrossRef Li X, Lian Z, Wang S, Xing L, Yu J. Interactions between EGFR and PD-1/PD-L1 pathway: implications for treatment of NSCLC. Cancer Lett. 2018;418:1–9.PubMedCrossRef
79.
Zurück zum Zitat Petrelli F, Maltese M, Tomasello G, Conti B, Borgonovo K, Cabiddu M, et al. Clinical and molecular predictors of PD-L1 expression in non-small-cell lung Cancer: systematic review and meta-analysis. Clin Lung Cancer. 2018;19:315–22.PubMedCrossRef Petrelli F, Maltese M, Tomasello G, Conti B, Borgonovo K, Cabiddu M, et al. Clinical and molecular predictors of PD-L1 expression in non-small-cell lung Cancer: systematic review and meta-analysis. Clin Lung Cancer. 2018;19:315–22.PubMedCrossRef
80.
Zurück zum Zitat Lan B, Ma C, Zhang C, Chai S, Wang P, Ding L, et al. Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: a meta-analysis. Oncotarget. 2018;9:7684–99.PubMedPubMedCentral Lan B, Ma C, Zhang C, Chai S, Wang P, Ding L, et al. Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: a meta-analysis. Oncotarget. 2018;9:7684–99.PubMedPubMedCentral
81.
Zurück zum Zitat Coelho MA, de Carne TS, Rana S, Zecchin D, Moore C, Molina-Arcas M, et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity. 2017;47:1083–99. e6PubMedPubMedCentralCrossRef Coelho MA, de Carne TS, Rana S, Zecchin D, Moore C, Molina-Arcas M, et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity. 2017;47:1083–99. e6PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Cha YJ, Shim HS. PD-L1 expression and CD8+ tumor-infiltrating lymphocytes are associated with ALK rearrangement and clinicopathological features in inflammatory myofibroblastic tumors. Oncotarget. 2017;8:89465–74.PubMedPubMedCentralCrossRef Cha YJ, Shim HS. PD-L1 expression and CD8+ tumor-infiltrating lymphocytes are associated with ALK rearrangement and clinicopathological features in inflammatory myofibroblastic tumors. Oncotarget. 2017;8:89465–74.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Boichard A, Tsigelny IF, Kurzrock R. High expression of PD-1 ligands is associated with kataegis mutational signature and APOBEC3 alterations. Oncoimmunology. 2017;6:e1284719.PubMedPubMedCentralCrossRef Boichard A, Tsigelny IF, Kurzrock R. High expression of PD-1 ligands is associated with kataegis mutational signature and APOBEC3 alterations. Oncoimmunology. 2017;6:e1284719.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Gadducci A, Guerrieri ME. Immune checkpoint inhibitors in gynecological cancers: update of literature and perspectives of clinical research. Anticancer Res. 2017;37:5955–65.PubMed Gadducci A, Guerrieri ME. Immune checkpoint inhibitors in gynecological cancers: update of literature and perspectives of clinical research. Anticancer Res. 2017;37:5955–65.PubMed
85.
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
86.
87.
Zurück zum Zitat Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9.PubMedPubMedCentralCrossRef Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103.PubMedCrossRef Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103.PubMedCrossRef
89.
Zurück zum Zitat Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–7.PubMedCrossRef Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–7.PubMedCrossRef
90.
Zurück zum Zitat Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45:931–43.PubMedCrossRef Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45:931–43.PubMedCrossRef
91.
Zurück zum Zitat Blacher E, Levy M, Tatirovsky E, Elinav E. Microbiome-modulated metabolites at the Interface of host immunity. J Immunol. 2017;198:572–80.PubMedCrossRef Blacher E, Levy M, Tatirovsky E, Elinav E. Microbiome-modulated metabolites at the Interface of host immunity. J Immunol. 2017;198:572–80.PubMedCrossRef
92.
Zurück zum Zitat Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84.PubMedPubMedCentralCrossRef Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Krieg C, Nowicka M, Guglietta S, Schindler S, Hartmann FJ, Weber LM, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24:144–53.PubMedCrossRef Krieg C, Nowicka M, Guglietta S, Schindler S, Hartmann FJ, Weber LM, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24:144–53.PubMedCrossRef
95.
Zurück zum Zitat Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.PubMedPubMedCentralCrossRef Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Fujisawa Y, Yoshino K, Otsuka A, Funakoshi T, Fujimura T, Yamamoto Y, et al. Baseline neutrophil to lymphocyte ratio combined with serum LDH level associated with outcome of nivolumab immunotherapy in a Japanese advanced melanoma population. Br J Dermatol. 2018; https://doi.org/10.1111/bjd.16427. Fujisawa Y, Yoshino K, Otsuka A, Funakoshi T, Fujimura T, Yamamoto Y, et al. Baseline neutrophil to lymphocyte ratio combined with serum LDH level associated with outcome of nivolumab immunotherapy in a Japanese advanced melanoma population. Br J Dermatol. 2018; https://​doi.​org/​10.​1111/​bjd.​16427.
97.
Zurück zum Zitat Weide B, Martens A, Hassel JC, Berking C, Postow MA, Bisschop K, et al. Baseline biomarkers for outcome of melanoma patients treated with Pembrolizumab. Clin Cancer Res. 2016;22:5487–96.PubMedPubMedCentralCrossRef Weide B, Martens A, Hassel JC, Berking C, Postow MA, Bisschop K, et al. Baseline biomarkers for outcome of melanoma patients treated with Pembrolizumab. Clin Cancer Res. 2016;22:5487–96.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Cabel L, Riva F, Servois V, Livartowski A, Daniel C, Rampanou A, et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann Oncol. 2017;28:1996–2001.PubMedCrossRef Cabel L, Riva F, Servois V, Livartowski A, Daniel C, Rampanou A, et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann Oncol. 2017;28:1996–2001.PubMedCrossRef
99.
Zurück zum Zitat Iijima Y, Hirotsu Y, Amemiya K, Higashi S, Miyashita Y, Omata M. Rapid decrease of circulating tumor DNA predicted the treatment effect of nivolumab in a lung cancer patient within only 5 days. Respir Med Case Rep. 2017;22:31–3.PubMedPubMedCentral Iijima Y, Hirotsu Y, Amemiya K, Higashi S, Miyashita Y, Omata M. Rapid decrease of circulating tumor DNA predicted the treatment effect of nivolumab in a lung cancer patient within only 5 days. Respir Med Case Rep. 2017;22:31–3.PubMedPubMedCentral
100.
Zurück zum Zitat Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, et al. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42.PubMedPubMedCentralCrossRef Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, et al. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Yue C, Jiang Y, Li P, Wang Y, Xue J, Li N, et al. Dynamic change of PD-L1 expression on circulating tumor cells in advanced solid tumor patients undergoing PD-1 blockade therapy. OncoImmunology. 2018; e1438111 Yue C, Jiang Y, Li P, Wang Y, Xue J, Li N, et al. Dynamic change of PD-L1 expression on circulating tumor cells in advanced solid tumor patients undergoing PD-1 blockade therapy. OncoImmunology. 2018; e1438111
102.
Zurück zum Zitat Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, et al. Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res. 2017;5:480–92.PubMedPubMedCentralCrossRef Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, et al. Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res. 2017;5:480–92.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Kruger S, Legenstein ML, Rosgen V, Haas M, Modest DP, Westphalen CB, et al. Serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death ligand 1 (sPD-L1) in advanced pancreatic cancer. Oncoimmunology. 2017;6:e1310358.PubMedPubMedCentralCrossRef Kruger S, Legenstein ML, Rosgen V, Haas M, Modest DP, Westphalen CB, et al. Serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death ligand 1 (sPD-L1) in advanced pancreatic cancer. Oncoimmunology. 2017;6:e1310358.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Nagato T, Ohkuri T, Ohara K, Hirata Y, Kishibe K, Komabayashi Y, et al. Programmed death-ligand 1 and its soluble form are highly expressed in nasal natural killer/T-cell lymphoma: a potential rationale for immunotherapy. Cancer Immunol Immunother. 2017;66:877–90.PubMedCrossRef Nagato T, Ohkuri T, Ohara K, Hirata Y, Kishibe K, Komabayashi Y, et al. Programmed death-ligand 1 and its soluble form are highly expressed in nasal natural killer/T-cell lymphoma: a potential rationale for immunotherapy. Cancer Immunol Immunother. 2017;66:877–90.PubMedCrossRef
105.
Zurück zum Zitat Caponnetto S, Iannantuono GM, Barchiesi G, Magri V, Gelibter A, Cortesi E. Prolactin as a potential early predictive factor in metastatic non-small cell lung Cancer patients treated with Nivolumab. Oncology. 2017;93:62–6.PubMedCrossRef Caponnetto S, Iannantuono GM, Barchiesi G, Magri V, Gelibter A, Cortesi E. Prolactin as a potential early predictive factor in metastatic non-small cell lung Cancer patients treated with Nivolumab. Oncology. 2017;93:62–6.PubMedCrossRef
106.
Zurück zum Zitat Yamazaki N, Kiyohara Y, Uhara H, Iizuka H, Uehara J, Otsuka F, et al. Cytokine biomarkers to predict antitumor responses to nivolumab suggested in a phase 2 study for advanced melanoma. Cancer Sci. 2017;108:1022–31.PubMedPubMedCentralCrossRef Yamazaki N, Kiyohara Y, Uhara H, Iizuka H, Uehara J, Otsuka F, et al. Cytokine biomarkers to predict antitumor responses to nivolumab suggested in a phase 2 study for advanced melanoma. Cancer Sci. 2017;108:1022–31.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.PubMedPubMedCentralCrossRef Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Dirkx AE, Oude Egbrink MG, Castermans K, van der Schaft DW, Thijssen VL, Dings RP, et al. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J. 2006;20:621–30.PubMedCrossRef Dirkx AE, Oude Egbrink MG, Castermans K, van der Schaft DW, Thijssen VL, Dings RP, et al. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J. 2006;20:621–30.PubMedCrossRef
109.
Zurück zum Zitat Atkins MB, Plimack ER, Puzanov I, Fishman MN, McDermott DF, Cho DC, et al. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. Lancet Oncol. 2018;19:405–15.PubMedCrossRef Atkins MB, Plimack ER, Puzanov I, Fishman MN, McDermott DF, Cho DC, et al. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. Lancet Oncol. 2018;19:405–15.PubMedCrossRef
110.
Zurück zum Zitat McKendry RT, Spalluto CM, Burke H, Nicholas B, Cellura D, Al-Shamkhani A, et al. Dysregulation of antiviral function of CD8(+) T cells in the chronic obstructive pulmonary disease lung. Role of the PD-1-PD-L1 Axis. Am J Respir Crit Care Med. 2016;193:642–51.PubMedPubMedCentralCrossRef McKendry RT, Spalluto CM, Burke H, Nicholas B, Cellura D, Al-Shamkhani A, et al. Dysregulation of antiviral function of CD8(+) T cells in the chronic obstructive pulmonary disease lung. Role of the PD-1-PD-L1 Axis. Am J Respir Crit Care Med. 2016;193:642–51.PubMedPubMedCentralCrossRef
111.
113.
Zurück zum Zitat Faruki H, Mayhew GM, Serody JS, Hayes DN, Perou CM, Lai-Goldman M. Lung adenocarcinoma and squamous cell carcinoma gene expression subtypes demonstrate significant differences in tumor immune landscape. J Thorac Oncol. 2017;12:943–53.PubMedCrossRef Faruki H, Mayhew GM, Serody JS, Hayes DN, Perou CM, Lai-Goldman M. Lung adenocarcinoma and squamous cell carcinoma gene expression subtypes demonstrate significant differences in tumor immune landscape. J Thorac Oncol. 2017;12:943–53.PubMedCrossRef
114.
Zurück zum Zitat Cortellini A, Bersanelli M, Buti S, Gambale E, Atzori F, Zoratto F, et al. Family history of cancer as surrogate predictor for immunotherapy with anti-PD1/PD-L1 agents: preliminary report of the FAMI-L1 study. Immunotherapy. 2018;10:643–55.PubMedCrossRef Cortellini A, Bersanelli M, Buti S, Gambale E, Atzori F, Zoratto F, et al. Family history of cancer as surrogate predictor for immunotherapy with anti-PD1/PD-L1 agents: preliminary report of the FAMI-L1 study. Immunotherapy. 2018;10:643–55.PubMedCrossRef
115.
Zurück zum Zitat Sun C, Mezzadra R, Schumacher TN. Regulation and function of the PD-L1 checkpoint. Immunity. 2018;48:434–52.PubMedCrossRef Sun C, Mezzadra R, Schumacher TN. Regulation and function of the PD-L1 checkpoint. Immunity. 2018;48:434–52.PubMedCrossRef
116.
Zurück zum Zitat Yamazaki N, Takenouchi T, Fujimoto M, Ihn H, Uchi H, Inozume T, et al. Phase 1b study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in Japanese patients with advanced melanoma (KEYNOTE-041). Cancer Chemother Pharmacol. 2017;79:651–60.PubMedPubMedCentralCrossRef Yamazaki N, Takenouchi T, Fujimoto M, Ihn H, Uchi H, Inozume T, et al. Phase 1b study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in Japanese patients with advanced melanoma (KEYNOTE-041). Cancer Chemother Pharmacol. 2017;79:651–60.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, et al. Pembrolizumab for platinum- and Cetuximab-refractory head and neck Cancer: results from a single-arm. Phase II Study J Clin Oncol. 2017;35:1542–9.PubMedCrossRef Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, et al. Pembrolizumab for platinum- and Cetuximab-refractory head and neck Cancer: results from a single-arm. Phase II Study J Clin Oncol. 2017;35:1542–9.PubMedCrossRef
118.
Zurück zum Zitat Hui R, Garon EB, Goldman JW, Leighl NB, Hellmann MD, Patnaik A, et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann Oncol. 2017;28:874–81.PubMedCrossRef Hui R, Garon EB, Goldman JW, Leighl NB, Hellmann MD, Patnaik A, et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann Oncol. 2017;28:874–81.PubMedCrossRef
119.
Zurück zum Zitat Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, et al. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody Pembrolizumab in melanoma. J Clin Oncol. 2016;34:4102–9.PubMedPubMedCentralCrossRef Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, et al. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody Pembrolizumab in melanoma. J Clin Oncol. 2016;34:4102–9.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung Cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35:3924–33.PubMedPubMedCentralCrossRef Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung Cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35:3924–33.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18:312–22.PubMedCrossRef Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18:312–22.PubMedCrossRef
122.
Zurück zum Zitat Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–13.PubMedPubMedCentralCrossRef Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–13.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16:257–65.PubMedPubMedCentralCrossRef Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16:257–65.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33:1430–7.PubMedCrossRef Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33:1430–7.PubMedCrossRef
125.
Zurück zum Zitat Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–30.PubMedCrossRef Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–30.PubMedCrossRef
126.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255–65.PubMedCrossRef Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255–65.PubMedCrossRef
128.
Zurück zum Zitat Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389:67–76.PubMedCrossRef Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389:67–76.PubMedCrossRef
129.
Zurück zum Zitat McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, et al. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol. 2016;34:833–42.PubMedCrossRef McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, et al. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol. 2016;34:833–42.PubMedCrossRef
130.
Zurück zum Zitat Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7.PubMedPubMedCentralCrossRef Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387:1837–46.PubMedCrossRef Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387:1837–46.PubMedCrossRef
132.
Zurück zum Zitat Powles T, O'Donnell PH, Massard C, Arkenau HT, Friedlander TW, Hoimes CJ, et al. Efficacy and safety of Durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3:e172411.PubMedPubMedCentralCrossRef Powles T, O'Donnell PH, Massard C, Arkenau HT, Friedlander TW, Hoimes CJ, et al. Efficacy and safety of Durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3:e172411.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Apolo AB, Infante JR, Balmanoukian A, Patel MR, Wang D, Kelly K, et al. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter. Phase Ib Study J Clin Oncol. 2017;35:2117–24.PubMedCrossRef Apolo AB, Infante JR, Balmanoukian A, Patel MR, Wang D, Kelly K, et al. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter. Phase Ib Study J Clin Oncol. 2017;35:2117–24.PubMedCrossRef
134.
Zurück zum Zitat Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–8.PubMedCrossRef Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–8.PubMedCrossRef
Metadaten
Titel
Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors
verfasst von
Ming Yi
Dechao Jiao
Hanxiao Xu
Qian Liu
Weiheng Zhao
Xinwei Han
Kongming Wu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0864-3

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Update Onkologie

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