Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2016

Open Access 01.12.2016 | Review

Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy

verfasst von: Weijie Ma, Barbara M. Gilligan, Jianda Yuan, Tianhong Li

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2016

Abstract

Modulating immune inhibitory pathways has been a major recent breakthrough in cancer treatment. Checkpoint blockade antibodies targeting cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programed cell-death protein 1 (PD-1) have demonstrated acceptable toxicity, promising clinical responses, durable disease control, and improved survival in some patients with advanced melanoma, non-small cell lung cancer (NSCLC), and other tumor types. About 20 % of advanced NSCLC patients and 30 % of advanced melanoma patients experience tumor responses from checkpoint blockade monotherapy, with better clinical responses seen with the combination of anti-PD-1 and anti-CTLA-4 antibodies. Given the power of these new therapies, it is important to understand the complex and dynamic nature of host immune responses and the regulation of additional molecules in the tumor microenvironment and normal organs in response to the checkpoint blockade therapies. In this era of precision oncology, there remains a largely unmet need to identify the patients who are most likely to benefit from immunotherapy, to optimize the monitoring assays for tumor-specific immune responses, to develop strategies to improve clinical efficacy, and to identify biomarkers so that immune-related adverse events can be avoided. At this time, PD-L1 immunohistochemistry (IHC) staining using 22C3 antibody is the only FDA-approved companion diagnostic for patients with NSCLC-treated pembrolizumab, but more are expected to come to market. We here summarize the current knowledge, clinical efficacy, potential immune biomarkers, and associated assays for immune checkpoint blockade therapies in advanced solid tumors.

Background

Since 2011, when the US Food and Drug Administration (FDA) approved ipilimumab for advanced melanoma, there has been an explosion of research interest and drug development for harnessing the immune system to fight against cancer. A general search for “immune therapy and cancer” in the clinicaltrials.gov database from 2000 to 2010 shows 640 hits, while the same search on March 20, 2015 shows 7815 hits and December 23, 2015 shows 8941 hits. Active research in tumor immunology includes studies on adoptive T cell therapy and cancer vaccine, as well as clinical investigations into immune checkpoint blockade in monotherapy or combination therapies. First generation programed cell-death protein 1 (PD-1) blockade antibodies pembrolizumab and nivolumab obtained US FDA approval for advanced melanoma in 2014 and as second line therapy for non-small cell lung cancer (NSCLC) in 2015. We have seen a surge in the availability of immune checkpoint inhibitors for multiple cancer indications. Forecasts for 2013–2020 project that the market for immunotherapy will increase from approximately $1 billion (US dollars) in 2013 to in excess of $7 billion in 2020 (corresponding to 33 % annual growth), across the seven major markets (USA, France, Germany, Italy, Spain, UK, and Japan) [1].
Unlike chemotherapy and molecularly targeted therapy, the checkpoint blockade immunotherapies result in durable clinical responses through the induction, activation, and expansion of tumor-specific cytotoxic T cells. Immune checkpoints play an essential role in maintaining self-tolerance and regulating the amplitude and duration of T cell responses. Two cytotoxic T cell immune checkpoint receptors (cytotoxic T-lymphocyte antigen 4 (CTLA-4) and PD-1) deliver inhibitory signals when bound to their respective ligands CD80/86 and programed cell-death ligand 1 (PD-L1)/2. Immunotherapies with checkpoint blockade antibodies that block CTLA-4 and PD-1 (or its ligand PD-L1) can restore and augment cytotoxic T cell responses against chemotherapy-refractory tumors, leading to durable responses and prolonged overall survival with tolerable toxicity. In the lymphatic tissues, anti-CTLA-4 antibody induces non-tumor-specific immune activation that is largely de novo induction of new responses. In tumor microenvironment, anti-PD-1/PD-L1 antibody targets tumor-induced immune defects and repairing ongoing tumor immunity. In addition to these anti-CTLA-4 and anti-PD-1/PD-L1 antibodies, drugs targeting other immune checkpoints and/or co-stimulatory ligand-receptor inhibitors, such as lymphocyte activation gene 3 protein (LAG3) and T cell immunoglobulin and mucin-domain containing protein 3 (TIM-3), are currently being explored in many tumor types and will be discussed further in the “Principles of cancer immunotherapy” section.
Given the distinct and novel mechanisms of action of immunotherapy on effector immune cells, rather than tumor cells, there are emerging tumor response patterns that are not accurately assessed by Response Evaluation Criteria in Solid Tumors (RECIST). In 2009, a guideline for immune-related response criteria (irRC) was proposed for patients with advanced melanoma receiving ipilimumab [2]. This guideline has been used to evaluate the tumor response for ongoing immune checkpoint blockade therapies [3]. When assessed appropriately, approximately 20 % of advanced NSCLC patients and 30–40 % of advanced melanoma patients have objective tumor responses to PD-1 blockade monotherapy. The combination of anti-PD-1 and anti-CTLA-4 antibodies increased the clinical response in advanced melanoma patients with a recent FDA approval [4]. In an attempt to increase the therapeutic, in checkpoint inhibitors, a companion diagnostic of PD-L1 immunohistochemistry (IHC) had been approved by the US FDA for selecting NSCLC patients treated with pembrolizumab. Interestingly, overall tumor responses, progression-free survival (PFS) and overall survival (OS) were not consistently associated with PD-L1 expression in the patients receiving immune checkpoint inhibitors [5]. In this age of personalized medicine, finding and validating better immune biomarkers will enable us to select patients for cancer immunotherapies. It will also lead to a better understanding of the unique mechanisms of tumor response and resistance, to allow for the development of strategies to improve clinical efficacy, and to lead to better evaluation of response and monitoring assays for host immune function. Validating potential immune biomarkers will enable us to better select patients for cancer immunotherapies and to avoid immune-related adverse effects (irAEs). Many excellent review articles on cancer biomarkers and immune checkpoint inhibitors have been recently published [612]. In this review, we will focus on the current knowledge, clinical efficacy, available monitoring assays, and emerging novel technologies for identifying translational biomarkers for immune checkpoint blockade immunotherapies in advanced solid tumors.

Principles of cancer immunotherapy

Clinically apparent cancer can be thought of as a host’s inability to eliminate transformed cells. Cancer immunotherapy refers to a diverse range of therapeutic approaches that harness the immune system to induce or restore the capacity of cytotoxic T cells, and other immune effector cells, and to recognize and eliminate cancer [13, 14]. The field of cancer immunotherapy is undergoing a renaissance due to a greater understanding of the immune system and immunosurveillance. The immune system protects the host against tumor development but also promotes tumor growth by selecting for tumors of lower immunogenicity. This dual effect of the immune system on developing tumors creates a dynamic process termed cancer immunoediting, which is comprised of three phases: elimination, equilibrium, and escape [15]. Cancer cells are immunogenic through the generation of tumor-specific mutant antigens (TSMA, neoantigens) from somatic gene structural and/or epigenetic alterations [16]. During the elimination phase, immune effector cells (mainly T and natural killer (NK) lymphocytes) are activated by the inflammatory cytokines released by the growing tumor cells, macrophages, and stromal cells surrounding the tumor cells. The recruited tumor-infiltrating NK cells and macrophages produce interleukin 2 (IL-2), interleukin 12 (IL-12), and interferon gamma (IFNγ), which kill tumor cells by cytotoxic mechanisms such as perforin, tumor necrosis factors (TNF)-related apoptosis-inducing ligands (TRAILs), and reactive oxygen species. Due to heterogeneity, tumor cells with a less immunogenic phenotype are able to escape this elimination phase (i.e., immunosurveillance) and expand during the equilibrium phase. The equilibrium phase may occur over a period of many years when, under Darwinian selection, new tumor cell variants emerge with mutations that further increase overall resistance to immune attack. During the escape phase, tumor cell variants breach the host’s immune defenses conferring further resistance to immune detection [17].
Major mechanisms for escape include defective antigen presentation, tumor-induced inhibitory checkpoint pathways against effector T cell activity, infiltrating immunosuppressive immune cells including regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs), and secretion of immunosuppressive cytokines (transforming growth factor beta (TGF-β), IL-6, vascular endothelial growth factor (VEGF)) [7, 18, 19]. Full activation of T and NK lymphocytes requires the coordinated participation of several surface receptors that meet their cognate ligands through structured transient cell-to-cell interactions known as immune synapses. Current PD-1/PD-L1 blockade therapies aim to boost the patient’s effector T cells to specifically recognize and kill cancer cells. Table 1 summarizes the current list of major immune checkpoint inhibitors that have obtained FDA approval or are in the late phases of clinical development.
Table 1
Summary of clinical indication and ongoing evaluation of immune checkpoint inhibitors in major cancer types
Target
Drug
Class
Company
Clinical indication and ongoing evaluation (status; approval date; trial identifier; country)
CTLA-4
Ipilimumab (Yervoy®, MDX-010, MDX-101)
Human IgG1/kappa
Bristol-Myers Squibb
Metastatic melanoma (US FDA approved on March 25, 2011); metastatic NSCLC (phase I has been completed; NCT01165216; Japan; phase II reported, NCT00527735, USA; phase III ongoing, NCT01285609; USA; phase III ongoing NCT02279732; China)
Tremelimumab (ticilimumab, CP-675206)
Human anti-CTLA-4 IgG2 mab
MedImmune/AstraZeneca
Metastatic melanoma (phase I has been completed; NCT01103635; USA; phase II has been completed, NCT00471887, USA); advanced hepatocellular carcinoma (phase II has been completed; NCT01008358; Spain); Metastatic NSCLC (phase Ib has been reported, NCT02000947, USA; phase II has been reported, NCT02179671, USA; first line, phase III MYSTIC ongoing; NCT02453282; USA)
PD-1
Nivolumab (Opdivo®, ONO-4538, MDX-1106, BMS-936558)
Human IgG4/kappa
Bristol-Myers Squibb; Ono Pharmaceuticals
Metastatic melanoma (Japan approval on July 4, 2014; US FDA accelerated approval on December 22, 2014; US FDA approval of nivolumab in combination with ipilimumab for BRAF V600 wild-type tumor on September 30, 2015); Squamous NSCLC (US FDA approval on March 4, 2015; European Commission on July 20, 2015); expands to non-squamous NSCLC (US FDA approval on October 9, 2015); advanced (metastatic) renal cell carcinoma (US FDA approval on November 23, 2015); classical Hodgkin lymphoma that has relapsed or progressed after autologous hematopoietic stem cell transplantation and post-transplantation brentuximab vedotin (US FDA approval on May 17, 2016)
Pembrolizumab (Keytruda®, lambrolizumab, MK-3475)
Humanized IgG4
Merck & Co.
Metastatic melanoma (USA accelerated approval on September 4, 2014 for patients with disease progression after ipilimumab and, if BRAF V600 mutation positive, a BRAF inhibitor; US FDA expanded to initial treatment on December 18, 2015); metastatic NSCLC whose tumors express PD-L1 as determined by an FDA-approved test and who have disease progression on or after platinum-containing chemotherapy (US FDA approval on October 2, 2015)
Pidilizumab (CT-011)
Humanized IgG1
CureTech Ltd
Diffuse large-B cell lymphoma (phase II has been completed; NCT00532259; USA); metastatic melanoma (phase II has been completed; NCT01435369; USA)
AMP-514 (MEDI0680)
Humanized IgG4
MedImmune
Advanced malignancies (phase II is currently recruiting participants; NCT02013804; USA)
AUNP-12
Peptide antagonist
Aurigene, Pierre Fabre
Cancer (preclinical phase, Aurigene granted Pierre Fabre worldwide rights to develop AUNP12 for cancer indications; announced on February 11, 2014; India)
PD-L1
BMS936559 (MDX-1105)
Human IgG4
Bristol-Myers Squibb
Advanced or recurrent solid tumors (phase II has been completed; NCT00729664; USA)
Atezolizumab (Tecentriq™, MPDL3280A, RG7446)
Human IgG1
Roche & Genentech
Metastatic bladder cancer (phase III, US FDA granted breakthrough therapy designation on May 31, 2014; priority review on March 14, 2016; accelerated approval on May 18, 2016); metastatic NSCLC (phase III, US FDA grants breakthrough therapy designation on February 1, 2015)
Durvalumab (MEDI4736)
Humanized IgG1
AstraZeneca
Glioblastoma (phase II is currently recruiting participants; NCT02336165; USA); metastatic squamous cell carcinoma of the head and neck (phase II is currently recruiting participants; NCT02207530; USA); advanced or metastatic NSCLC (phase III is currently recruiting participants; NCT02352948; Global study); advanced colorectal cancer (phase II is currently recruiting participants; NCT02227667; USA); metastatic NSCLC (first line phase III MYSTIC is current recruiting participants; NCT02453282; USA; first line phase III ARCTIC is current recruiting participants; NCT02352948; Global); metastatic bladder cancer (US FDA granted breakthrough therapy designation for PD-L1-positive tumors in patients who progressed during or after one standard platinum-based regimen on February 17, 2016)
Avelumab (MSB0010718C)
Fully humanized IgG1
Merck KGaA, EMD Serono, Pfizer
Advanced solid tumors (phase I with consecutive parallel group expansion; currently recruiting participants in multiple tumor types and settings; NCT01772004; USA); metastatic NSCLC (phase III is currently recruiting participants after failure of a platinum-based doublet; NCT02395172; and first line versus platinum doublet; NCT02576574; USA)
PD-L2
AMP-224
PD-L2-IgG2a fusion protein
Amplimmune
Advanced cancer (phase II has been completed; NCT01352884; USA)
Last assessed information at ClinicalTrial.gov on December 28, 2015; updated FDA approvals on May 18, 2016. Italicized data highlights major cancer types in clinical evaluation
Abbreviations: CTLA-4 cytotoxic T-lymphocyte-associated protein 4, NSCLC non-small-cell lung cancer, PD1 programed cell-death protein 1, PDL1 programed cell-death ligand 1
Immune checkpoints normally maintain self-tolerance, regulate the amplitude and duration of T cell responses, and prevent tissue damage. PD-1 and PD-L1 immune checkpoint blockade can restore the function of exhausted CD8 T cells in chronic viral infection, augmenting the existing tumor-specific immunity [20, 21]. Nevertheless, immune checkpoint inhibitors can override immune self-tolerance, inducing a unique syndrome of autoimmune and autoinflammatory side effects (i.e., irAEs) [22]. Many of these irAEs resemble autoimmune diseases, such as autoinflammatory rheumatic disease, dermatologic disease, colitis, hepatitis, and endocrinopathies [23]. Therefore, almost all immunotherapy clinical trials have excluded individuals with preexisting autoimmune diseases, although a recent study demonstrates that the use of ipilimumab in cancer patients with autoimmune diseases is safe and effective [24]. However, a recent study showed the safety and efficacy of using anti-CTLA-4 antibody in patients with autoimmune disease [24]. Early detection and effective management of these irAEs are pivotal for a favorable clinical outcome. Theoretically, cytotoxic T cells against tumors and normal organs are different subclones that may not need to be activated in parallel. Further studies are warranted to specifically enhance tumor-specific cytotoxic T cell response and avoid unnecessary T cell response to normal tissues.

Biology and mechanisms of PD-1 and PD-L1 molecules

PD-1 (CD279) is expressed on many cell types including T cells, B cells, natural killer T cells, activated monocytes, and dendritic cells (DCs) in humans and mice [25]. In normal human lymphoid tissues, PD-1 is detected on germinal center-associated T cells [26]. PD-1 binds to the ligand PD-L1 (B7-H1, CD274) or PD-L2 (B7-DC, CD272). PD-L1 is typically expressed on the surface of tumor cells [27]. PD-L1 is also constitutively expressed on other cell types such as T and B cells, DCs, macrophages, mesenchymal stem cells, and bone marrow-derived mast cells [28]. Cell surface expression of PD-L1 can be upregulated on both tumor cells and other cell types after treatment with type I or type II interferons (IFNs) [29, 30], radiation [3133], or chemotherapy [3438]. Additionally, radiotherapy may induce direct tumor-cell killing and multiple immunomodulatory changes that can potentially influence the effectiveness of immunotherapy [3133].
Engagement of PD-1 by its ligands, either PD-L1 or PD-L2, induces a negative control signal resulting in the inhibition of T cell proliferation, cytokine production, and cytotoxic activity. PD-L1 can provide inhibitory signals to activated T cells through interactions with the surface receptor PD-1 or CD80 [25, 39]. These signals enable PD-L1-expressing tumor cells to evade immune detection by NK cells or T cells [4042]. PD-L2 is encoded by PDCD1LG2, which is essential for T cell proliferation and IFN production. PD-L2 can stimulate PD-1 receptor signaling, resulting in “T cell exhaustion,” a temporary inhibition of activation and proliferation that can be reversed by the removal of PD-1 signal [25].
Upregulation of PD-L1 on different tumor types inhibits the local antitumor T cell response. Figure 1 illustrates the expression and complex interaction of key molecules and co-stimulatory ligand receptors between tumor and immune cells in tumor microenvironment (TME), and the two potential mechanisms proposed to understand the biology and function of PD-L1 on tumor cells [43]. While constitutive oncogenic signals (such as protein kinase B (AKT), epidermal growth factor receptor (EGFR), KRAS pathways) upregulate PD-L1 expression on tumors as innate (tumor cell intrinsic) resistance (Fig. 1, left) [4448], inflammatory signals generate cytokine-induced PD-L1 expression on either tumor cells or immune cells (myeloid suppressor cells, dendritic cell, macrophage, and lymphocytes) in the tumor microenvironment as adaptive resistance (Fig. 1, right) [49, 50]. Preclinical studies have demonstrated that blocking the PD-L1/PD-1 interaction increases the numbers of effector T cells, augments cytolytic activity of tumor-specific T cells, enhances the production of pro-inflammatory cytokines, brings effector T cells to the tumor sites, reduces numbers and suppression of Tregs at the tumor site, and downregulates the production of suppressive cytokines IL-10 [5154]. Tumor-infiltrating T cells may also be functionally inert, due in part to the expression of PD-1 along with other inhibitory receptors [27, 55]. The hallmarks of the PD-1/PD-L1 blockage effect include immune modulation at tumor site, direct targeting of tumor-induced immune defects, and repairing ongoing tumor immunity. PD-1 deletion in mice can lead to autoimmunity, most notably when bred onto backgrounds of autoimmune-susceptible mouse strains [56, 57]. In multiple syngeneic mouse tumor models, blockade of PD-1 or its ligands promotes antitumor activity [58, 59]; anti-PD-1 activity in vivo can be enhanced by combination with antibodies to other T cell-negative regulators, such as CTLA-4 and LAG-3 [6062]. A recent study suggests that PD-L1 is an independent prognostic marker in melanoma, defining a tumor subset with distinct genetic and morphophenotypic features [63].

Clinical efficacy of PD-1/PD-L1 blockade therapies and PD-L1 expression

Nivolumab and pembrolizumab are the first generation PD-1 immune checkpoint blockade antibodies. Nivolumab inhibits the interaction between PD-1 and its ligands, PD-L1, and PD-L2 with similar IC50 values (2.52 and 2.59 nmol/L, respectively). Nivolumab enhances T cell reactivity in the presence of a T cell receptor stimulus at the low concentration (~1.5 ng/mL) without nonspecific lymphocyte activation. Nivolumab also enhances the production of inflammatory cytokines in assays such as the human T cell/DC-mixed lymphocyte reaction (MLR) assays, and Staphylococcal enterotoxin B (SEB), and cytomegalovirus (CMV) recall response assays. In addition, nivolumab increases antigen-specific CD8+ T cell responses in patients with melanoma [64]. Nivolumab completely restored CD4+ T-responder cell proliferation and partially restored IFNγ production. Nivolumab also overcomes Treg suppression of CD8+ T cells by increasing resistance to Treg suppression and also by directly limiting suppressive capacity of Treg [65]. This was demonstrated in monkeys when increased numbers of CD8+ T-effector memory cells were detected in CMV-positive peripheral blood after repeated treatment at the highest dose of nivolumab for 3 months. Similarly, marked accumulation of CD8+ effector memory T cells in lymphoid organs and tissues of PD-1-deficient mice has been described [66].
Both nivolumab and pembrolizumab have shown significant survival benefit in some patients with advanced melanoma, NSCLC, and renal cell carcinomas in early clinical trials (Table 1). These agents were generally well tolerated, even in elderly patients with prolonged dosing. Lower tumor response rates have been observed in never smokers and patients with EGFR mutations or ALK rearrangements when compared to heavy smokers. Table 2 summarizes this clinical efficacy data from four of the most studied PD-1 and PD-L1 inhibitors at various stages of clinical development. As only subsets of patients are benefiting from the PD-1/PD-L1 immune checkpoint blockade therapies, it is important to try to select for this population so that patients less likely to improve with therapy can be spared toxicities. An additional challenge is that response criteria used for standard therapy may not be applicable for immunotherapy. Unlike standard therapeutics in which tumor response is measured by a variety of radiographic and laboratory tests every 6–8 weeks, delayed tumor responses due to the time required for activation and function of effector T cells are frequently observed in cancer patients treated with immune checkpoint blockade therapies. In some patients, tumor regression persisted after discontinuation of PD-1 blockade therapy [67]. Evaluating responses appropriately and identifying patients who are most likely to respond are important aspects of immunotherapy that continue to be developed.
Table 2
Summary of reported clinical efficacy of PD-1/PD-L1 inhibitors
Agent
Clinical trials identifier
Phase of clinical trial
Sample size (no. Pt)
Patient population
Biomarker
Regimen
Tumor responses (ORR)
Median PFS (months)
OS (months; median unless otherwise specified)
Reference: author (year)
Nivolumab
NCT01642004 (CheckMate 017)
Phase III
272 all: 135 nivolumab, 137 docetaxel
Advanced squamous NSCLC with disease progression during or after first-line chemotherapy
PD-L1-positive tumor cells
Nivolumab 3 mg/kg IV every 2 weeks
20 %
3.5
9.2
Brahmer J (2015) [125]
Docetaxel 75 mg/m2 IV every 3 weeks
9 %
2.8
6
NCT01673867 (CheckMate 057)
Phase III
582 all: 292 nivolumab, 290 Docetaxel
Advanced non-squamous NSCLC after platinum-based doublet chemotherapy
PD-L1-positive tumor cells
Nivolumab 3 mg/kg IV every 2 weeks
19 %
2.3
12.2
Borghaei H (2015) [126]
Docetaxel 75 mg/m2 IV every 3 weeks
12 %
4.2
9.4
NCT01668784 (CheckMate 025)
Phase III
821 all: 406 nivolumab, 397 Everolimus
Advanced clear-cell renal-cell carcinoma with one or two regimens of anti-angiogenic therapy
PD-L1-positive tumor cells
Nivolumab 3 mg/kg IV every 2 weeks
25 %
4.6
25
Motzer RJ (2015) [127]
Everolimus 10 mg orally daily
5 %
4.4
19.6
NCT01721746 (CheckMate 037)
Phase III
631 all: 272 nivolumab,133 investigators choice of chemo
Unresectable or metastatic melanoma after ipilimumab or ipilimumab and BRAF inhibitor if BRAF positive
PD-L1-positive tumor cells
Nivolumab 3 mg/kg IV every 2 weeks
32 %
4.7
NA
Weber JS (2015) [128]
Chemo: either dacarbazine 1000 mg/m2 IV every 3 weeks or carboplatin AUC = 6 plus paclitaxel 185 mg/m2 IV every 3 weeks
11 %
4.2
NA
NCT01927419 (CheckMate 069)
Phase III
142 all: 95 nivolumab + ipilimumab, 47 ipilimumab
Unresectable or metastatic melanoma treatment naïve with measurable disease
Tissue available for PD-L1 biomarker analysis
Nivolumab 1 mg/kg IV every 3 weeks × 4 doses plus ipilimumab 3 mg/kg IV every 3 weeks × 4 doses, then maintenance nivolumab 3 mg/kg IV every 2 weeks
BRAF wild type: 61 %; BRAF mutation: 52 %
BRAF wild type: NR; BRAF mutation: 8.5
NA
Postow MA (2015) [4]
Same dose schedule with nivolumab placebo in both the combination and maintenance phase
BRAF wild type: 11 %; BRAF mutation: 22 %
BRAF wild type: 4.4; BRAF mutation: 2.7
NA
NCT01721772 (CheckMate 066)
Phase III
418 all: 210 nivolumab, 208 dacarbazine
Untreated metastatic melanoma without BRAF mutation
Tissue available for PD-L1 biomarker analysis
Nivolumab 3 mg/kg IV every 2 weeks plus placebo every 3 weeks
40 %
5.1
1-year OS: 72.9 %
Robert C (2015) [129]
Dacarbazine 1000 mg/m2 IV every 3 weeks plus placebo every 2 weeks
13.9 %
2.2
1-year OS: 42.1 %
NCT01844505 (CheckMate 067)
Phase III
945 all: 316 nivolumab, 314 combination, 315 ipilimumab
Untreated, unresectable stage III or IV melanoma
Tissue available for PD-L1 biomarker analysis
Nivolumab 3 mg/kg IV every 2 weeks (plus ipilimumab placebo)
43.7 %
6.9
NA
Larkin J (2015) [130]
Nivolumab 1 mg/kg IV every 3 weeks plus ipilimumab 3 mg/kg IV every 3 weeks × 4 doses; then maintenance nivolumab 3 mg/kg IV every 2 weeks
58 %
11.5
Ipilimumab 3 mg/kg IV every 3 weeks (plus nivolumab placebo)
19 %
2.9
NCT01721759 (CheckMate 063)
Phase II single arm trial
117
Advanced NSCLC
PD-L1-positive tumor cells
Nivolumab 3 mg/kg every 2 weeks until progression or unacceptable toxic effects
14.5 %
1.9
8.2
Rizvi NA (2015) [131]
NCT00730639
Phase I with expansion cohorts
107
Advanced melanoma
Unselected
Nivolumab at 1, 3, or 10 mg/kg every 2 weeks for up to 96 weeks
31 %
3.7
16.8
Topalian SL (2014) [67]
Phase I with expansion cohorts
34
Previously treated advanced RCC
Unselected
Nivolumab at 1, 3, or 10 mg/kg every 2 weeks for up to 96 weeks
29 %
7.3
22.4
McDermott DF (2015) [132]
Phase II with expansion cohorts
129
Heavily pretreated advanced NSCLC
Unselected
Nivolumab at 1, 3, or 10 mg/kg every 2 weeks for up to 96 weeks
17 %
2.6
9.9
Gettinger SN (2015) [133]
Pembrolizumab
NCT01866319 (KEYNOTE-006)
Phase III
834 all: 279 pembrolizumab, 277 pembrolizumab, 278 ipilimumab
Unresectable stage III or IV melanoma
PD-L1-positive tumor cells
Pembrolizumab 3 mg/kg IV every 2 weeks
33.7 %
5.5
NA
Robert C (2015) [134]
Pembrolizumab 3 mg/kg IV every 3 weeks
32.9 %
4.1
Ipilimumab 3 mg/kg IV every 3 weeks
11.9 %
2.8
NCT01704287 (KEYNOTE-002)
Phase II
540 all: 180 pembrolizumab, 181 pembrolizumab, 179 chemotherapy
Ipilimumab-refractory melanoma
Will be reported with the final overall survival analysis
Arm A 2 mg/kg (n = 180)
21 %
5.4
NA
Ribas A (2015) [135]
Arm B 10 mg/kg (n = 181)
25 %
5.8
Chemotherapy
4 %
3.6
NCT012958297 (KEYNOTE-001)
Phase I
495
Advanced NSCLC
PD-L1-positive tumor cells
Pembrolizumab 2 or 10 mg/kg IV every 3 weeks or 10 mg/kg every 2 weeks over a 30-min period
19.4 %
3.7
12
Garon EB (2015) [5]
Phase I
655
Melanoma
Unselected
Pembrolizumab 2 mg/kg every 3 weeks (Q3W), 10 mg/kg Q3W, or 10 mg/kg Q2W until unacceptable toxicity, disease progression, or investigator decision
33 %
12-month PFS 35 %
23
Adil Daud (2015) [136]; Ribas A (2016) [137]
Phase I with expansion cohort
173
Advanced melanoma after at least 2 ipilimumab doses
Unselected
Pembrolizumab 2 mg/kg IV every 3 week or 10 mg/kg IV every 3 weeks
26 %
2
NA
Robert C (2014) [138]
NCT01848834 (KEYNOTE-012)
Phase IB
32
Metastatic triple-negative breast cancer
PDL-1-positive tumor cells
Pembrolizumab 10 mg/kg IV every 2 weeks
19 %
6-month PFS 23.3 %
NA
Nanda R (2014) [139]
NCT1905657 (KEYNOTE-010)
Phase II/III
1034 all: 339 pembrolizumab, 343 pembrolizumab, 309 docetaxel
Previously treated PD-L1-positive advanced NSCLC
PDL-1-positive tumor cells
Pembrolizumab 2 mg/kg, IV every 3 weeks
18 %
3.9 months
14.9
Herbst RS [2015] [140]
Pembrolizumab 10 mg/kg, IV every 3 weeks
18.5 %
4.0 month
17.3
Docetaxel, 75 mg/m2 every 3 weeks
9.3 %
4.0 month
8.2
NCT01953692 (KEYNOTE-013)
Phase IB
15
Hodgkin lymphoma
Unselected
Pembrolizumab 10 mg/kg IV every 2 weeks up to 2 years
53 %
NA
NA
Moskowitz C (2014) [141]
Atezolizumab (MPDL3280A)
NCT01846416
Phase II
205
NSCLC
PD-L1-positive tumor cells
Atezolizumab 1200 mg IV every 3 weeks
The highest ORR was seen in pts with PD-L1 TC3 or IC3 tumors
NA
NA
Spigel DR (2015) [142]
NCT01903993 (POPLAR)
Phase II
287
Previously treated NSCLC patients (pts) were stratified by PD-L1 IC status
PD-L1-positive tumor cells
Atezolizumab 1200 mg IV every 3 weeks
57 %
2.7
12.6
Spira AI (2015) [143]; Fehrenbacher L (2016)
Docetaxel 75 mg/m2 IV every 3 weeks
24 %
3.0
9.7
NCT01375842
Phase I
35
Metastatic melanoma
PD-L1-positive tumor cells
Atezolizumab IV every 3 weeks for up to 1 year
26 %
24-week PFS 35 %
NA
Omid Hamid (2013) [144]
Phase I
277
Multiple cancer types
PD-L1-positive tumor cells
Atezolizumab intravenously every 3 weeks doses >1 ml/kg
18 %
2.6
NA
Herbst RS (2014) [145]
NCT01633970
Phase Ib
37
Untreated NSCLC
PD-L1-positive tumor cells
Atezolizumab 15 mg/kg IV every 3 weeks with standard chemo dosing for 4–6 cycles followed by MPDL3280A maintenance therapy until progression
67 %
NA
NA
Stephen V (2015) [146]
Phase Ib
14
Arm A: refractory metastatic colorectal cancer; arm B: oxaliplatin-naive mCRC
Not mentioned
Arm A: MPDL3280A 20 mg/kg every 3 weeks and bevacizumab (bev) 15 mg/kg every 3 weeks
8 % (1/13) in arm A
NA
NA
Bendell, J.C. (2015) [147]
Arm B: MPDL3280A 14 mg/kg every 2 weeks, bev 10 mg/kg every 2 weeks and mFOLFOX6 at standard doses
36 % (9/25) in Arm B
NA
NA
Phase Ib
12
Metastatic renal cell carcinoma
Not selected
Atezolizumab 15 mg/kg given alone on cycle 1 day 1 and concurrently with 20 mg/kg every 2 weeks thereafter
40 %
NA
NA
Sznol M (2015) [148]
Durvalumab (MEDI4736)
NCT01693562
Phase I/II
198
NSCLC
Tissue available for PD-L1 biomarker analysis
Durvalumab 10 mg/kg IV every 2 weeks until unacceptable toxicity, disease progression, or for up to 12 months
14 % (23 % in PD-L1+ tumors)
NA
NA
Rizvi NA (2015) [149]
Phase I
13
NSCLC
Tissue available for PD-L1 biomarker analysis
Durvalumab 7 doses (1–25) across 6 cohorts (0.1–10 mg/kg every 2 weeks; 15 mg/kg every 3 weeks)
NA
NA
NA
Brahmer JR (2014) [150]
Multi-arm expansion study
62
A squamous cell carcinoma of the head and neck expansion cohort
Tissue available for PD-L1 biomarker analysis
Durvalumab IV every 2 weeks at 10 mg/kg for 12 months
12 % (25 % in PD-L1+ pts)
NA
NA
Segal NH (2015) [151]
Phase I
26
Advanced solid tumors
Durvalumab IV every 2 (q2w) or 3 weeks (q3w) in a 3 + 3 dose escalation with a 28-day (q2w) or 42-day (q3w) DLT window
NA
NA
NA
Lutzky J (2014) [152]
NCT02088112
Phase I
10
NSCLC
Unselect
Durvalumab cohort A received 3 mg/kg (starting dose) every 2 weeks plus gefitinib 250 mg QD
NA
NA
NA
Creelan BC (2015) [153]
NCT02000947
Phase Ib
118 (102 eligible)
NSCLC
Tissue available for PD-L1 biomarker analysis
Durvalumab 10–20 mg/kg every 2 or 4 weeks plus tremelimumab 1 mg/kg (N = 56)
23 % (6/26): 22 % (2/9) in PD-L1+ versus 29 % (4/14) in PD-L1-
NA
NA
Antonia SJ (2015) [154]; Updated in Antonia SJ (2016) [155]
Durvalumab 10–20 mg/kg every 2 weeks plus tremelimumab 3 mg/kg (N = 34)
20 % (5/25)
Durvalumab 15 mg/kg every 4 weeks plus tremelimumab 10 mg/kg (N = 9)
0 % (0/9)
Abbreviations: DLT dose-limiting toxicity, q2w every 2 weeks, q3w every 3 weeks, q4w every 4 weeks, QD once daily, BRAF B-raf and v-raf murine sarcoma viral oncogene homologue B1, NA not available

Biomarkers for PD-1/PD-L1 blockade therapies

Translational biomarker research is critical for the future of drug development for clinical immune checkpoint blockade therapy. Only one PD-L1 IHC has been approved as a companion diagnostic by the US FDA for patients with advanced NSCLC for pembrolizumab [68]. The challenges in developing immuno-oncology biomarkers for clinical use have been recognized by regulatory agencies and professional societies such as the Society for Immunotherapy of Cancer (SITC) [69]. Over the past several years, the SITC Immune Biomarkers Task force has provided the roadmap and regular updates on immune biomarker development [7072]. Immunological changes in peripheral blood and tumor could potentially reflect tumor response in patients and serve as immune biomarkers. The immune monitoring assays had been developed to evaluate these potential biomarkers before and after immune checkpoint blockade therapies and investigate their correlation with clinical outcome (Fig. 2). These biomarkers could be classified into two groups: tumor-derived immune biomarkers and immune cell-derived biomarkers as discussed in the following sections. Table 3 summarizes currently available quantitative biomarker assays for immune checkpoint inhibitors.
Table 3
Currently available translational biomarker assays for immune checkpoint inhibitors
Biospecimen
Method
Tissue/cell types
Pros
Cons
References and recommended reading
FFPE
IHC
Tumor cells or tumor infiltrating immune cells
Direct detection; accurately pinpoint cancer cells; highly sensitive; simplicity; low cost
Requirement of trained pathologists; inconsistency for criteria used to score tumors such as PD-L1-positive or negative
Herbst R (2014) [145]; Loughlin PA (2007) [164]
Multicolor IHC
Tumor cells or tumor infiltrating immune cells
Broad dynamic range; capability for multiplexing using different fluorescence channels; >10 protein targets are identified in the same sample; amenability for co-localization studies
Absence of rigorous quantitative tests; limitation in some biomarker-driven clinical trials; user must select combinations of dyes
Carvajal-Hausdorf DE (2014) [165]
T cell receptor deep sequencing
TILs
T cell count information;
T cell clonality in tumor
Heterogeneous expression of TIL
Robbins HS (2013) [100]
Whole exome sequencing (WES)
Tumor cells
Characterization of tumor mutation load including nucleotide substitutions; structural rearrangements and copy number alterations; identification of the neoantigens and neoepitopes; affordable cost
Require high-performance deep sequencing, computational bioinformatics support;
The pipelines are still at early developmental phase
Snyder A (2014) [104]; Rizvi NA (2015) [105];
Bouffet E (2016) [107];
Chen K-H (2016) [166]
Hugo W (2016) [106]
Blood
ELISPOT assays (IFNγ and granzyme B)
T cells in PBMCs
Detection of tumor antigen-specific CD4+ and CD8+ T cell response with good assay sensitivity;
Relatively well validated assay
A poor correlation with clinically relevant immune responses
Shafer-Weaver K (2006) [167]; Janetzki S (2008) [95] Malyguine A (2012) [94]; Janetzki S (2015) [93];
Flow cytometry (tetramer, polyfunctional analysis)
T cells in PBMCs
Assessment of tumor antigen-specific CD4+ and CD8+ T cells response; measure multiple functions; detection of neoantigen-specific CD8 + PD-1+ T cells; minimally invasive
Merely in lab research, not as routine clinical monitoring yet
Yuan J (2008) [84]; Attic S (2011) [85]; McNeil LK (2013) [86]; Barrera L (2015) [168]; Gros A (2016) [118]
Flow cytometry phenotype staining
Whole blood immune phenotype
Analyses of the frequency and proliferation of different subsets of immune cells; routine operation
Dedicated resource and staff to perform the analyses
Streitz M (2013) [169]; van Dongen JJ (2012) [170]
RNA-Seq (NGS)
T cells in PBMCs
Identification of genetic variants; a broader dynamic range; detection of more differentially expressed genes; fast and high efficiency
More expensive than microarray; more complex for analysis; bulk signature, not single cell signals; need more validation
Zhao S (2010) [171]
qPCR assay
T cells in PBMCs
High specificity; able to detect the reactivity of low-frequency T cells in the peripheral blood of metastatic cancer patients
Bulk signature, not single cell signals; need more validation
Kammula US (2008) [172]
Flow cytometry
CTCs
Qualitative analysis at the single cell level in a relatively short period of time; decrease the amount of blood needed; provide valuable information regarding the frequency, phenotype and/or the functionality of T cells
Expensive; need more validation
Zaritskaya L (2010) [83]
Cell sieve microfiltration assay and QUASR technique
CTCs
PD-L1 levels from CTCs or CAMLs serves as a surrogate for PD-L1 expression in tumor; as a marker for immunotherapy response
Limited in lab research; need more validation
Steven HL (2015) [173]; Adams DL (2014) [174]
Abbreviations: FFPE formalin-fixed paraffin-embedded, PD-L1 program death-1, TIL tumor-infiltrating lymphocytes, IHC immunohistochemistry, CAMLs cancer-associated macrophage-like cells, ELISPOT enzyme-linked immunospot assay, CTL cytotoxic T-lymphocytes, CTCs circulating tumor cells, PBMC peripheral blood mononuclear cell, WES whole exome sequencing, NGS next-generation sequencing

Current immune monitoring assays

IHC staining

PD-L1 is the best studied immuno-oncology biomarker to date. Currently, PD-L1 IHC using 22C3 antibody is the only FDA-approved companion diagnostic for selecting NSCLC patients for pembrolizumab [68]. Table 4 summarizes the IHC assays used for PD-L1 expression using baseline archival tumor specimens in these clinical studies [73]. There are many variables in these IHC assays. First, the time between sample collection and treatment with a PD-1 or PD-L1 inhibitor is not controlled. Second, tumor cells, immune cells, as well as stroma cells can express PD-L1 with considerably heterogeneity within the tumor microenvironment. Third, PD-L1 expression is induced by IFNγ during disease progression and treatment. Fourth, different PD-L1 antibodies were used for different immune checkpoint blockade clinical studies. Currently, there is no validated antibody for IHC staining for this class of immune checkpoint inhibitors—each sponsor uses different antibodies. Antibodies staining tumor cells or stroma cells may have different PD-L1 epitopes (Table 4). An additional challenge is that none of the commercial PD-L1 IHC test has been validated in terms of clinical outcomes. Head-to-head comparisons among three different commercially available PD-L1 assays (Dako 22C3, Dako 28-8, and Ventana SP263 as the reference) using 500 formalin-fixed, paraffin-embedded archival samples of NSCLC tissue obtained from commercial sources found 91 to 95 % correlation, which is comparable to typical within assay agreement for IHC across the dynamic range at multiple cutoffs [74]. A recent meta-analysis demonstrates that PD-L1 expression is significantly associated with clinical response to anti-PD-1/PD-L1 antibodies in patients with malignant melanoma (MM) or non-squamous NSCLC. However, a proportion of PD-L1-negative patients also benefits from anti-PD-1 therapy in MM and squamous-NSCLC. Thus, expression of PD-L1 in tumor tissues cannot be used as a predictive biomarker of eligibility for treatment with anti-PD-1/PD-L1 antibodies. It may represent a correlation marker for non-squamous NSCLC and melanoma [75].
Table 4
Immunohistochemistry assays for PD-L1 expression
Drug
Antibody (marker)
Rx line
Tumor type
Targeted cells
Tumor specimen
Cutoff point (%)
ORR % in IHC+ cases (95 % CI)
ORR % in IHC− cases (95 % CI)
Predictive role
P value
References
Nivolumab
28-8 rabbit (Dako)
1 L
Melanoma
TCs
Archival FFPE or new biopsy
5
58 (46,69)
41 (35,48)
Yes
0.001
Larkin J (2015) [130]
Nivolumab + ipilimumab
72 (60,82)
55 (48,62)
Yes
0.001
Nivolumab
≥2 L
5
44 (30,58)
20 (11,32)
Yes
NA
Weber JS (2015) [128]
Nivolumab + ipilimumab
1 L
5
58 (37,78)
55 (42,69)
No
>0.05
Postow MA (2015) [4]
Nivolumab
1 L
5
53 (41,64)
33 (25,42)
No
>0.05
Robert C (2015) [129]
≥2 L
NSCLC
TCs
Archival FFPE or new biopsy
1
31 (23,40)
9 (5,16)
Yes
0.002
Paz-Ares L (2015) [156]; Updated in Borghaei H (2015) [126]
5
36 (26,46)
10 (6,17)
Yes
0.002
10
37 (27,48)
11 (6,17)
Yes
0.002
≥2 L
Archival FFPE
1
17 (9,29)
17 (8,29)
No
0.9364
Brahmer JR (2015) [125]
5
21 (10,37)
15 (8,25)
No
0.2908
10
19 (8,36)
16 (9,26)
No
0.6411
≥2 L
Archival FFPE
1
20 (5,35)
13 (2,28)
No
>0.05
Rizvi NA (2015) [131]
5
24 (5,43)
14 (3,25)
No
1 L
Archival FFPE
5
31 (NA)
10 (NA)
No
>0.05
Gettinger SN (2015) [157]
Nivolumab + ipilimumab
1 L
Archival FFPE
5
19 (NA)
14 (NA)
No
>0.05
Antonia SJ (2014) [158]
Nivolumab
5H1 and anti-PD-1 monoclonal M3
≥2 L
Archival FFPE
5
39 (34,44)
6 (1,12)
Yes
0.025
Taube JM (2014) [113]
Pembrolizumab
22C3 mouse (Dako)
≥1 L
NSCLC
TCs and ICs
New biopsy
50
45 (33,57)
17 (10,25)
Yes
0.001
Garon EB (2015) [5]
1 L
New biopsy
50
47 (23,72)
19 (8,38)
Yes
NA
Rival NA (2015) [159]
Any
Archival FFPE
1
25 (NA)
13 (NA)
Yes
NA
Garon EB (2014) [160]
≥1 L
Archival FFPE
50
30 (23,39)
9.8 (NA)
Yes
NA
Herbst RS (2015) [140]
29 (22,37)
10.7 (NA)
1 L
Archival FFPE
50
16 (NA)
10 (NA)
Yes
NA
Garon EB (2014) [161]
Atezolizumab (MPDL3280A)
SP142 rabbit (Roche Ventana)
≥2 L
NSCLC
TCs and ICs
Archival FFPE and new biopsy
50
45 (23,68)
14 (6,25)
Yes
NA
Leora H (2015) [162]
≥2 L
NSCLC
1+
31 (25,37)
20 (14,26)
Yes
0.015
Herbst RS (2014) [145]
Solid Tumor
1+
29 (27,31)
13 (10,16)
Yes
0.007
≥2 L
NSCLC
2+
18 (NA)
8 (NA)
Yes
NA
Spira AI (2015) [143]
Durvalumab (MEDI-4736)
SP263 rabbit (Roche Ventana)
≥2 L
NSCLC
TCs
Archival FFPE and new biopsy
25
39 (NA)
5 (NA)
Yes
NA
Segal NH (2014) [163]
33 (13,59)
30 (16,47)
No for combo
NA
Antonia SJ (2016) [155];
22 (3, 60)
29 (8, 58)
No for monotherapy
NA
NA not available
The presence of tumor-infiltrating lymphocytes (TILs) at tumor microenvironment has been associated with an improved clinical outcome [7678], regardless of cancer treatment. TIL counts are primarily measured by IHC [79]. Recent development of multicolored immunohistochemistry staining resulted in a multiplex of up to seven fluorescent dyes effectively interrogated in microscopes equipped with a multispectral camera [80]. The multispectral fluorescent IHC with a panel including CD3, CD8, FoxP3, CD163, and PD-L1 was used to quantify the density of TIL subpopulation in advanced melanoma patients.

Flow cytometry staining

Flow cytometry traditionally uses fluorochrome-labeled antibodies to identify cellular protein expression of the target cells. Flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) and TILs can quantitate the effect of therapy on low-frequency immune subsets such as Treg, MDSC, and activated CD8+ T cells [81]. The biggest advantage of flow cytometry is the combination of multiparameter measurements in a high-throughput manner. With recent advances in laser and fluorochrome technology, commercially available systems allow for detection of up to 18 colors at analysis rates of more than 20,000 events per second. This tremendous power allows for the analysis of multiple characteristics of rare cells such as tumor antigen-specific T-lymphocytes, including their phenotype and functional activity [82]. Flow cytometric assays are currently widely used for the enumeration and phenotypic characterization of lymphocytes, measuring cytokines, secreted molecules, intracellular signaling, function, and cell proliferation [83]. Using polychromatic flow cytometry, multiple surface and intracellular markers can be detected, allowing in-depth characterization of T cell phenotype and activation state [84]. In addition to flow cytometry, antigen-loaded soluble major histocompatibility complex (MHC) tetramer stains can be used to analyze tumor-specific T cell subpopulations [82]. This tetramer/pentamer staining has been used for phenotypic and quantitative analysis of antigen-specific CD8 T cells [85, 86]. The human leukocyte antigen (HLA) system is a gene complex encoding the MHC proteins in humans. HLA-peptide complexes binding to T cell receptors could further define the specificity for epitopes and alleles [87]. Recently, mass cytometry or CyTOF which replace the fluorescent labels with heavy metal ions is under investigation for its application in immune biomarker research [88].

ELISA and protein microarray

Biomarkers in the blood have been pursued because specimens are easy to collect and assays are available for serial monitoring and quantitative measurements [89]. Enzyme-linked immunoabsorbent assay (ELISA) is widely used to assess the production of soluble mediators such as cytokines, chemokines, and tumor antigen-specific antibodies in peripheral blood before and after treatment. Novel protein microarray technologies such as ProtoArray® are available to analyze the serological response of up to 9000 proteins simultaneously [9092].

ELISPOT

Enzyme-linked immunospot (ELISPOT) assays are robust and standardized tests that quantify both the frequency and function of T or B cells at the single cell level. Both PBMCs and isolated CD4+ and CD8+ T cell subsets could be stimulated by antigens and cultured in a 96-well plate with a nitrocellulose membrane coated with antibodies for cytokines of interest. These assays, such as the IFNγ ELISPOT, have gained increasing popularity for monitoring clinical trials and in basic research [9395]. Results from various clinical trials, including peptide and whole tumor cell vaccination and cytokine treatment, showed the suitability of the IFNγ ELISPOT assay for monitoring T cell response [96]. ELISPOT assays revealed that 75 % of melanoma patients vaccinated with antigenic peptide demonstrated specific immune responses [97, 98]. Besides IFNγ, other cytokines such as IL-2, IL-5, IL-10, IL-17, granzyme B, TNF, and granulocyte-macrophage colony-stimulating factor (GM-CSF) are commonly measured by ELISPOT assays. It has been expanded to more than 2~3 parameters with the introduction of fluorescent dyes [99].

T cell or B cell receptor deep sequencing

To develop a robust, quantitative method with high reproducibility, Robins et al. [100] uses a digital droplet PCR technique call a QuanTILfy assay. High-throughput quantitative sequencing of the rearranged TCR β genes uses the ImmunoSeq assay. The TCR loci undergo somatic gene rearrangements in the 52 variable (V), diversity (D), and 13 joining (J) regions of the β chain and variable (V) and joining (J) regions of α chain. There are 52 Vβ segments and 13 Jβ segments that can rearrange. With the right TaqMan probes-DNA from T cells can be easily identified with reference to total DNA, as was shown when purified human T cells were obtained from a healthy donor and then diluted and mixed with human lung fibroblasts at different ratios. This technique can also identify clonal expansion (the mark of the adaptive immune response). TCR deep sequencing has been used to show the correlation between TCR repertoire and clinical response to cancer immunotherapy in patients with melanoma or prostate cancer treated with ipilimumab [101]. Creation of a responder library for TCR repertoire will greatly advance the field, especially for genetically modified TCR cell transfer therapy.

WES for mutation load

The cancers that respond best to PD-1 inhibitors, such as advanced melanoma and NSCLC, are the tumor types that are genetically very complex with a high nonsynchronous mutation load. This discovery led to an interest in neoantigens (i.e., tumor-specific mutant antigens (TSMAs)) that are immunogenic [102]. The advances in both affordable next-generation sequencing (NGS) technology and bioinformatics make it feasible to assess the full mutation load in individual tumors and to identify neoantigens by comparing WGS of tumor and normal tissues [103]. Computation prediction algorithms and the tandem mini-gene library allow for the evaluation of the immunogenicity of both CD4+ and CD8+ T cell neoepitopes. Mutation load and neoepitopes have been explored for their correlation with clinical outcomes in cancer patients treated with the PD-1/PD-L1 immune checkpoint blockade [104107]. However, whole exome sequencing (WES) currently is not used in routine clinical practice. The role of tumor load assessed in targeted exome sequencing (TES) assays in predicting response to immune checkpoint inhibitors remains to be determined.

Genetic susceptibility

Genetic factors are known to modulate the immune response to a therapeutic protein product. In particular, some HLA haplotypes may predispose patients to developing either exceptional or undesirable antibody responses to immune checkpoint blockage therapies [108]. If both are appropriate and feasible, HLA mapping studies may help define a subset of patients who are at increased risk. Moreover, genetic polymorphisms in cytokine genes may upregulate or downregulate immune responses [109], which is why the FDA recommends that researchers evaluate the genetic factors that may modulate the immune response to a therapeutic protein. For example, the subset of patients who generate neutralizing antibodies to IFNβ products are more likely to possess distinct HLA haplotypes [108]. Thus, knowledge of the heightened susceptibility of patients with such HLA haplotypes may guide us to prevent such responses or pursuit other treatment options (http://​www.​fda.​gov/​ucm/​groups/​fdagov-public/​@fdagov-drugs-gen/​documents/​document/​ucm338856.​pdf).

Tumor-derived immune biomarkers

PD-L1 expression

Upregulation of PD-L1 expression level has been reported in many different cancer types (e.g., melanoma (40–100 %), NSCLC (35–95 %), and multiple myeloma (93 %)). High levels of PD-L1 expression have been linked to poor clinical outcomes [42, 110, 111]. Until now, anti-PD-L1 IHC on tumor specimens is the most commonly used biomarker for selecting patients who are likely to respond to treatments [89]. Constitutively overexpressed PD-L1 in melanoma is associated with poor clinical outcome [112]. However, the PD-L1 overexpression in the context of CD8-positive cells is associated with a better prognosis [113]. Although PD-L1 IHC using the 22C3 antibody has been approved by the US FDA as the only predictive companion diagnostic for selecting NSCLC patients for pembrolizumab, tumor responses have been seen in low PD-L1-expressing tumors. Conflicting results have been reported with other PD-L1 antibodies and drugs. Table 4 summarizes the reported data of PD-L1 IHC in current clinical trials.
Several tumor characteristics, such as a high mutation load, smoking-related tumors, and tumors with mismatch repair genes are associated with improved objective tumor response rate (ORR), durable clinical benefit (DCB), and PFS to immune checkpoint blockade antibodies. Notably, whole exome sequencing has revealed that a nonsynonymous mutation load of >200 per tumor correlates with clinical responses to PD-1 mAb in NSCLC, melanoma, and colorectal cancer with microsatellite instability (MSI; i.e., mutations in DNA mismatch repair genes) [114116]. Rizvi and his colleagues showed that mutation load correlates with improved ORR (63 versus 0 %), DCB (73 versus 13 %), and PFS (14.5 versus 3.7 months) [105]. Moreover, the efficacy was associated with a molecular smoking signature. The ORR was 56 and 17 % in transversion-high tumors and transversion-low tumors, respectively; the rate of DCB was 77 versus 22 %, respectively; and the PFS was also significantly longer in transversion-high tumors compared to transversion-low tumors. Efficacy was also connected with higher neoantigen burden, but additional genetic features are likely to play a role in determining the tumor response [116, 117]. Neoantigen-specific CD8+ T cell responses paralleled tumor regression in one responder, implying that anti-PD-1 therapy enhanced neoantigen-specific T cell reactivity [118].

Immune cell-derived biomarkers

TIL cells have been shown to express significantly higher levels of PD-1 than T cells in normal tissue [119]. The tumor microenvironment may secrete pro-inflammatory cytokines to upregulate the expression of PD-1 on TIL cells to ensure that they can respond to the high levels of PD-L1 expressed on the tumor [120]. Preexisting CD8+ T cells distinctly located at the invasive tumor margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis [121]. The proliferation of intratumoral CD8+ T cells directly correlated with radiographic reduction in tumor size in metastatic melanoma patients treated with pembrolizumab. Pretreatment samples obtained from responding patients showed higher numbers of CD8+ T cells, PD-1, and PD-L1-expressing cells at the invasive tumor margin and inside tumors, with a more clonal TCR repertoire. Using multivariate analysis, researchers established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Their findings indicate that tumor regression after PD-1 blockade requires preexisting CD8+ T cells that are negatively regulated by PD-1/PD-L1-mediated adaptive immune resistance. In addition, the tumor microenvironment can be dissected histopathologically in order to characterize spatial relationships between tumor and immune infiltrate, and transcriptional profiling assays can evaluate changes in gene expression in both the tumor and in lymphocytes [122].
Given that TIL in the tumor microenvironment affects the prognosis of the tumor, a new TNM Immune system (TNM-I) system has been proposed and is currently being validated by an international consortium [123]. Early studies suggest that activated T cells are not pharmacodynamic markers of nivolumab treatment [124]. It remains to be determined whether nivolumab treatment increases CD8 effector memory cells in cancer patients. With technical advances, detection of CD8-positive, PD-1-positive T cells in the peripheral blood of melanoma patients receiving PD-1 inhibitors is feasible. Furthermore, the tumor-antigen specificities and TCR repertoires of the circulating and tumor-infiltrating CD8+PD-1+ cells appeared similar [118].

Conclusions

The era of cancer immunotherapy has arrived. This major breakthrough in cancer treatment holds great promise for increasing cure rates for patients with various cancer types in multiple disease settings. Checkpoint blockade antibodies targeting CTLA-4, PD-1, and PD-L1 axis have demonstrated impressive and durable disease control and promising responses in patients with multiple tumor types. Despite these advances, only a subset of patients benefits from immune checkpoint blockade therapies, with some patients experiencing mechanism-based irAEs. Translational biomarker research is one way to overcome these limitations of therapy. Biomarkers play a critical role in understanding potential mechanisms of action in patients, identifying patients who are likely to respond to the growing list of cancer therapeutics and avoiding the irAEs. Several tumor-derived biomarkers have been reported from recent correlative studies including PD-L1 expression on tumor cells, high tumor mutational load, and neoantigens. In addition, several immune cell-derived biomarkers showed better correlation with the clinical outcomes. The presence of TILs in tumor microenvironments, increased PD-L1 expression on immune cells, and the ratio of effector CD8+ T cells to FoxP3+ regulatory T cells in tumors are some examples of markers for clinical outcomes. Future studies are warranted to harmonize companion diagnostics (such as PD-L1 IHC) for accurate clinical assessment and application of the class of PD-1/PD-L1 inhibitors. It is of importance to further evaluate mutation load as a potential biomarker and develop effective tumor antigen-specific T cell assays to differentiate immunogenic neoepitopes from putative ones. The advances in translational immune biomarker research are essential for personalized cancer immunotherapy as either monotherapy or combinational therapy.

Abbreviations

AKT, protein kinase B; APCs, antigen-presenting cells; BRAF, B-raf and v-raf murine sarcoma viral oncogene homologue B1; BTLA, B- and T-lymphocyte attenuator; CAMLs, cancer-associated macrophage-like cells; CEACAM, carcinoembryonic antigen-related cell adhesion molecule; CTCs, circulating tumor cells; CTL, cytotoxic T-lymphocytes; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; DC, dendritic cell; DLT, dose-limiting toxicity; EGFR, epidermal growth factor receptor; ELISPOT, enzyme-linked immunospot assay; FFPE, formalin-fixed paraffin-embedded; GITR, glucocorticoid-induced tumor necrosis factor receptor; GM-CSF, granulocyte-macrophage colony-stimulating factor; HLA, human leukocyte antigen; HVEM, herpes virus entry mediator; ICOS, inducible costimulators; IDO, indoleamin 2,3-dioxygenase; IHC, immunohistochemistry; IL-2R, IL-2 receptor; irAEs, immune-related adverse effects; JNK, c-Jun N-terminal kinase; LAG, lymphocyte-activation gene; MDSCs, myeloid-derived suppressor cells; MEK/ERK, mitogen/extracellular signal regulated kinase; MHC, major histocompatibility complex; NGS, next-generation sequencing; NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells; NSCLC, non-small-cell lung cancer; PBMCs, peripheral blood mononuclear cells; PD-1, programed death-1; PD-L1, programed cell-death ligand 1; PI3K, phosphatidylinositol 3-kinase; q2w, every 2 weeks; q3w, every 3 weeks; q4w, every 4 weeks; QD, once daily; STAT, signal transducer and activator of transcription; Teff, effector T cell; TGF-β, transforming growth factor beta; TIGIT, T cell immunoreceptor with Ig and ITIM domains; TIL, tumor-infiltrating lymphocytes; TIM-3, T cell immunoglobulin domain and mucin domain; TNF, tumor necrosis factors; Treg, regulatory T cells; TSMA, tumor-specific mutant antigen; VEGF, vascular endothelial growth factor; WES, whole exome sequencing

Acknowledgements

None.

Funding

None.

Availability of data and materials

The dataset supporting the conclusions of this article is included within the article.

Authors’ contributions

WM, JY, and TL contributed to the conception and design of the study. WM, JY, and TL carried out the development of methodology. WM, JY, and TL analyzed and interpreted the data. WM, BMG, JY, and TL wrote, reviewed, revised, and approved the final submission of the study. All authors read and approved the final manuscript.

Competing interests

JY owns a TCR sequencing patent with Adaptive Biotechnologies.
Not applicable.
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.

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 Webster RM. The immune checkpoint inhibitors: where are we now? Nat Rev Drug Discov. 2014;13(12):883–4.PubMedCrossRef Webster RM. The immune checkpoint inhibitors: where are we now? Nat Rev Drug Discov. 2014;13(12):883–4.PubMedCrossRef
2.
Zurück zum Zitat Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–20.PubMedCrossRef Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–20.PubMedCrossRef
3.
Zurück zum Zitat Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud A, Hamid O, et al. Evaluation of immune-related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab. J Clin Oncol. 2016;34(13):1510–7.PubMedCrossRef Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud A, Hamid O, et al. Evaluation of immune-related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab. J Clin Oncol. 2016;34(13):1510–7.PubMedCrossRef
4.
Zurück zum Zitat Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17.PubMedCrossRef Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17.PubMedCrossRef
5.
Zurück zum Zitat Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.PubMedCrossRef Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.PubMedCrossRef
7.
Zurück zum Zitat Pico de Coana Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med. 2015;21(8):482–91.PubMedCrossRef Pico de Coana Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med. 2015;21(8):482–91.PubMedCrossRef
8.
Zurück zum Zitat Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161(2):205–14.PubMedCrossRef Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161(2):205–14.PubMedCrossRef
10.
Zurück zum Zitat Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol. 2015;36(4):198–216.PubMedCrossRef Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol. 2015;36(4):198–216.PubMedCrossRef
11.
12.
Zurück zum Zitat Goel G, Sun W. Advances in the management of gastrointestinal cancers—an upcoming role of immune checkpoint blockade. J Hematol Oncol. 2015;8:86.PubMedPubMedCentralCrossRef Goel G, Sun W. Advances in the management of gastrointestinal cancers—an upcoming role of immune checkpoint blockade. J Hematol Oncol. 2015;8:86.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy—inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res. 2012;18(24):6580–7.PubMedCrossRef Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy—inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res. 2012;18(24):6580–7.PubMedCrossRef
15.
Zurück zum Zitat Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMedCrossRef Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMedCrossRef
16.
Zurück zum Zitat Hacohen N, Fritsch EF, Carter TA, Lander ES, Wu CJ. Getting personal with neoantigen-based therapeutic cancer vaccines. Cancer Immunol Res. 2013;1(1):11–5.PubMedPubMedCentralCrossRef Hacohen N, Fritsch EF, Carter TA, Lander ES, Wu CJ. Getting personal with neoantigen-based therapeutic cancer vaccines. Cancer Immunol Res. 2013;1(1):11–5.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Stewart TJ, Abrams SI. How tumours escape mass destruction. Oncogene. 2008;27(45):5894–903.PubMedCrossRef Stewart TJ, Abrams SI. How tumours escape mass destruction. Oncogene. 2008;27(45):5894–903.PubMedCrossRef
19.
Zurück zum Zitat Terme M, Pernot S, Marcheteau E, Sandoval F, Benhamouda N, Colussi O, et al. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013;73(2):539–49.PubMedCrossRef Terme M, Pernot S, Marcheteau E, Sandoval F, Benhamouda N, Colussi O, et al. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013;73(2):539–49.PubMedCrossRef
20.
Zurück zum Zitat Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439(7077):682–7.PubMedCrossRef Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439(7077):682–7.PubMedCrossRef
22.
Zurück zum Zitat Bertrand A, Kostine M, Barnetche T, Truchetet ME, Schaeverbeke T. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med. 2015;13:211.PubMedPubMedCentralCrossRef Bertrand A, Kostine M, Barnetche T, Truchetet ME, Schaeverbeke T. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med. 2015;13:211.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Corsello SM, Barnabei A, Marchetti P, De Vecchis L, Salvatori R, Torino F. Endocrine side effects induced by immune checkpoint inhibitors. J Clin Endocrinol Metab. 2013;98(4):1361–75.PubMedCrossRef Corsello SM, Barnabei A, Marchetti P, De Vecchis L, Salvatori R, Torino F. Endocrine side effects induced by immune checkpoint inhibitors. J Clin Endocrinol Metab. 2013;98(4):1361–75.PubMedCrossRef
24.
Zurück zum Zitat Johnson DB, Sullivan RJ, Ott PA, Carlino MS, Khushalani NI, Ye F, et al. Ipilimumab therapy in patients with advanced melanoma and preexisting autoimmune disorders. JAMA Oncol. 2016;2(2):234–40.PubMedCrossRef Johnson DB, Sullivan RJ, Ott PA, Carlino MS, Khushalani NI, Ye F, et al. Ipilimumab therapy in patients with advanced melanoma and preexisting autoimmune disorders. JAMA Oncol. 2016;2(2):234–40.PubMedCrossRef
25.
Zurück zum Zitat Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMedCrossRef Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMedCrossRef
26.
Zurück zum Zitat Dorfman DM, Brown JA, Shahsafaei A, Freeman GJ. Programmed death-1 (PD-1) is a marker of germinal center-associated T cells and angioimmunoblastic T-cell lymphoma. Am J Surg Pathol. 2006;30(7):802–10.PubMedPubMedCentralCrossRef Dorfman DM, Brown JA, Shahsafaei A, Freeman GJ. Programmed death-1 (PD-1) is a marker of germinal center-associated T cells and angioimmunoblastic T-cell lymphoma. Am J Surg Pathol. 2006;30(7):802–10.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015;21(1):24–33.PubMedPubMedCentralCrossRef Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015;21(1):24–33.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, et al. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol. 2002;169(10):5538–45.PubMedCrossRef Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, et al. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol. 2002;169(10):5538–45.PubMedCrossRef
29.
Zurück zum Zitat Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation. 2002;9(2):133–45.PubMedPubMedCentralCrossRef Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation. 2002;9(2):133–45.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Schreiner B, Mitsdoerffer M, Kieseier BC, Chen L, Hartung HP, Weller M, et al. Interferon-beta enhances monocyte and dendritic cell expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: relevance for the immune modulatory effect in multiple sclerosis. J Neuroimmunol. 2004;155(1-2):172–82.PubMedCrossRef Schreiner B, Mitsdoerffer M, Kieseier BC, Chen L, Hartung HP, Weller M, et al. Interferon-beta enhances monocyte and dendritic cell expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: relevance for the immune modulatory effect in multiple sclerosis. J Neuroimmunol. 2004;155(1-2):172–82.PubMedCrossRef
32.
Zurück zum Zitat Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–7.PubMedCrossRef Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–7.PubMedCrossRef
33.
Zurück zum Zitat Daly ME, Monjazeb AM, Kelly K. Clinical trials integrating immunotherapy and radiation for non-small-cell lung cancer. J Thorac Oncol. 2015;10(12):1685–93.PubMedCrossRef Daly ME, Monjazeb AM, Kelly K. Clinical trials integrating immunotherapy and radiation for non-small-cell lung cancer. J Thorac Oncol. 2015;10(12):1685–93.PubMedCrossRef
34.
Zurück zum Zitat Ramakrishnan R, Assudani D, Nagaraj S, Hunter T, Cho HI, Antonia S, et al. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest. 2010;120(4):1111–24.PubMedPubMedCentralCrossRef Ramakrishnan R, Assudani D, Nagaraj S, Hunter T, Cho HI, Antonia S, et al. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest. 2010;120(4):1111–24.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39(1):74–88.PubMedCrossRef Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39(1):74–88.PubMedCrossRef
36.
Zurück zum Zitat Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, Yang H, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729–41.PubMedCrossRef Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, Yang H, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729–41.PubMedCrossRef
37.
Zurück zum Zitat Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70(8):3052–61.PubMedCrossRef Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70(8):3052–61.PubMedCrossRef
38.
Zurück zum Zitat Kanterman J, Sade-Feldman M, Biton M, Ish-Shalom E, Lasry A, Goldshtein A, et al. Adverse immunoregulatory effects of 5FU and CPT11 chemotherapy on myeloid-derived suppressor cells and colorectal cancer outcomes. Cancer Res. 2014;74(21):6022–35.PubMedCrossRef Kanterman J, Sade-Feldman M, Biton M, Ish-Shalom E, Lasry A, Goldshtein A, et al. Adverse immunoregulatory effects of 5FU and CPT11 chemotherapy on myeloid-derived suppressor cells and colorectal cancer outcomes. Cancer Res. 2014;74(21):6022–35.PubMedCrossRef
39.
Zurück zum Zitat Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27(1):111–22.PubMedPubMedCentralCrossRef Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27(1):111–22.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Benson Jr DM, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116(13):2286–94.PubMedPubMedCentralCrossRef Benson Jr DM, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116(13):2286–94.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–7.PubMedPubMedCentralCrossRef Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–7.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296–304.PubMedCrossRef Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296–304.PubMedCrossRef
43.
Zurück zum Zitat Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.PubMedPubMedCentralCrossRef Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 2015;5(8):860–77.PubMedPubMedCentralCrossRef Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 2015;5(8):860–77.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Chen N, Fang W, Zhan J, Hong S, Tang Y, Kang S, et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol. 2015;10(6):910–23.PubMedCrossRef Chen N, Fang W, Zhan J, Hong S, Tang Y, Kang S, et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol. 2015;10(6):910–23.PubMedCrossRef
46.
Zurück zum Zitat Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol. 2014;25(10):1935–40.PubMedCrossRef Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol. 2014;25(10):1935–40.PubMedCrossRef
47.
Zurück zum Zitat Ota K, Azuma K, Kawahara A, Hattori S, Iwama E, Tanizaki J, et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin Cancer Res. 2015;21(17):4014–21.PubMedCrossRef Ota K, Azuma K, Kawahara A, Hattori S, Iwama E, Tanizaki J, et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin Cancer Res. 2015;21(17):4014–21.PubMedCrossRef
48.
Zurück zum Zitat Lastwika KJ, Wilson 3rd W, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76(2):227–38.PubMedCrossRef Lastwika KJ, Wilson 3rd W, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76(2):227–38.PubMedCrossRef
49.
Zurück zum Zitat Lou Y, Diao L, Parra Cuentas ER, Denning WL, Chen L, Fan YH, et al. Epithelial-mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res. 2016. doi:10.1158/1078-0432.CCR-15-1434. Lou Y, Diao L, Parra Cuentas ER, Denning WL, Chen L, Fan YH, et al. Epithelial-mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res. 2016. doi:10.​1158/​1078-0432.​CCR-15-1434.
51.
Zurück zum Zitat Spranger S, Koblish HK, Horton B, Scherle PA, Newton R, Gajewski TF. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014;2:3.PubMedPubMedCentralCrossRef Spranger S, Koblish HK, Horton B, Scherle PA, Newton R, Gajewski TF. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014;2:3.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Gatalica Z, Snyder C, Maney T, Ghazalpour A, Holterman DA, Xiao N, et al. Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev. 2014;23(12):2965–70.PubMedCrossRef Gatalica Z, Snyder C, Maney T, Ghazalpour A, Holterman DA, Xiao N, et al. Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev. 2014;23(12):2965–70.PubMedCrossRef
53.
Zurück zum Zitat Okiyama N, Katz SI. Programmed cell death 1 (PD-1) regulates the effector function of CD8 T cells via PD-L1 expressed on target keratinocytes. J Autoimmun. 2014;53:1–9.PubMedPubMedCentralCrossRef Okiyama N, Katz SI. Programmed cell death 1 (PD-1) regulates the effector function of CD8 T cells via PD-L1 expressed on target keratinocytes. J Autoimmun. 2014;53:1–9.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, Honjo T, et al. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer. 2010;116(7):1757–66.PubMedCrossRef Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, Honjo T, et al. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer. 2010;116(7):1757–66.PubMedCrossRef
55.
Zurück zum Zitat Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37.PubMedPubMedCentralCrossRef Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–51.PubMedCrossRef Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–51.PubMedCrossRef
57.
Zurück zum Zitat Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291(5502):319–22.PubMedCrossRef Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291(5502):319–22.PubMedCrossRef
58.
Zurück zum Zitat Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9(5):562–7.PubMedCrossRef Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9(5):562–7.PubMedCrossRef
59.
Zurück zum Zitat Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65(3):1089–96.PubMed Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65(3):1089–96.PubMed
60.
Zurück zum Zitat Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107(9):4275–80.PubMedPubMedCentralCrossRef Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107(9):4275–80.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedPubMedCentralCrossRef Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Massi D, Brusa D, Merelli B, Ciano M, Audrito V, Serra S, et al. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol. 2014;25(12):2433–42.PubMedCrossRef Massi D, Brusa D, Merelli B, Ciano M, Audrito V, Serra S, et al. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol. 2014;25(12):2433–42.PubMedCrossRef
64.
Zurück zum Zitat Wong RM, Scotland RR, Lau RL, Wang C, Korman AJ, Kast WM, et al. Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol. 2007;19(10):1223–34.PubMedCrossRef Wong RM, Scotland RR, Lau RL, Wang C, Korman AJ, Kast WM, et al. Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol. 2007;19(10):1223–34.PubMedCrossRef
65.
Zurück zum Zitat Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol. 2009;21(9):1065–77.PubMedPubMedCentralCrossRef Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol. 2009;21(9):1065–77.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Charlton JJ, Chatzidakis I, Tsoukatou D, Boumpas DT, Garinis GA, Mamalaki C. Programmed death-1 shapes memory phenotype CD8 T cell subsets in a cell-intrinsic manner. J Immunol. 2013;190(12):6104–14.PubMedCrossRef Charlton JJ, Chatzidakis I, Tsoukatou D, Boumpas DT, Garinis GA, Mamalaki C. Programmed death-1 shapes memory phenotype CD8 T cell subsets in a cell-intrinsic manner. J Immunol. 2013;190(12):6104–14.PubMedCrossRef
67.
Zurück zum Zitat Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30.PubMedPubMedCentralCrossRef Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, et al. Defining the critical hurdles in cancer immunotherapy. J Transl Med. 2011;9:214.PubMedPubMedCentralCrossRef Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, et al. Defining the critical hurdles in cancer immunotherapy. J Transl Med. 2011;9:214.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Butterfield LH, Disis ML, Khleif SN, Balwit JM, Marincola FM. Immuno-oncology biomarkers 2010 and beyond: perspectives from the iSBTc/SITC biomarker task force. J Transl Med. 2010;8:130.PubMedPubMedCentralCrossRef Butterfield LH, Disis ML, Khleif SN, Balwit JM, Marincola FM. Immuno-oncology biomarkers 2010 and beyond: perspectives from the iSBTc/SITC biomarker task force. J Transl Med. 2010;8:130.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Bedognetti D, Balwit JM, Wang E, Disis ML, Britten CM, Delogu LG, et al. SITC/iSBTc cancer immunotherapy biomarkers resource document: online resources and useful tools—a compass in the land of biomarker discovery. J Transl Med. 2011;9:155.PubMedPubMedCentralCrossRef Bedognetti D, Balwit JM, Wang E, Disis ML, Britten CM, Delogu LG, et al. SITC/iSBTc cancer immunotherapy biomarkers resource document: online resources and useful tools—a compass in the land of biomarker discovery. J Transl Med. 2011;9:155.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, et al. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer. 2016;4:3.PubMedPubMedCentralCrossRef Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, et al. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer. 2016;4:3.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Hansen AR, Siu LL. PD-L1 testing in cancer: challenges in companion diagnostic development. JAMA Oncol. 2016;2(1):15–6.PubMedCrossRef Hansen AR, Siu LL. PD-L1 testing in cancer: challenges in companion diagnostic development. JAMA Oncol. 2016;2(1):15–6.PubMedCrossRef
74.
Zurück zum Zitat Ratcliffe MJ, Sharpe A, Midha A, Barker C, Scorer P, Walker J. A comparative study of PD-L1 diagnostic assays and the classification of patients at PD-L1 positive and PD-L1 negative [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20. New Orleans, LA Philadelphia (PA): AACR; 2016. Abstract nr LB-094. Ratcliffe MJ, Sharpe A, Midha A, Barker C, Scorer P, Walker J. A comparative study of PD-L1 diagnostic assays and the classification of patients at PD-L1 positive and PD-L1 negative [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20. New Orleans, LA Philadelphia (PA): AACR; 2016. Abstract nr LB-094.
75.
Zurück zum Zitat Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016;100:88–98.PubMedCrossRef Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016;100:88–98.PubMedCrossRef
76.
Zurück zum Zitat Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, et al. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58(3):449–59.PubMedCrossRef Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, et al. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58(3):449–59.PubMedCrossRef
77.
Zurück zum Zitat Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol. 2012;124(2):192–8.PubMedPubMedCentralCrossRef Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol. 2012;124(2):192–8.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Erdag G, Schaefer JT, Smolkin ME, Deacon DH, Shea SM, Dengel LT, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72(5):1070–80.PubMedPubMedCentralCrossRef Erdag G, Schaefer JT, Smolkin ME, Deacon DH, Shea SM, Dengel LT, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72(5):1070–80.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Feng Z, Puri S, Moudgil T, Wood W, Hoyt CC, Wang C, et al. Multispectral imaging of formalin-fixed tissue predicts ability to generate tumor-infiltrating lymphocytes from melanoma. J Immunother Cancer. 2015;3:47.PubMedPubMedCentralCrossRef Feng Z, Puri S, Moudgil T, Wood W, Hoyt CC, Wang C, et al. Multispectral imaging of formalin-fixed tissue predicts ability to generate tumor-infiltrating lymphocytes from melanoma. J Immunother Cancer. 2015;3:47.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Stack EC, Wang C, Roman KA, Hoyt CC. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods. 2014;70(1):46–58.PubMedCrossRef Stack EC, Wang C, Roman KA, Hoyt CC. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods. 2014;70(1):46–58.PubMedCrossRef
81.
Zurück zum Zitat Ichihara F, Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin Cancer Res. 2003;9(12):4404–8.PubMed Ichihara F, Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin Cancer Res. 2003;9(12):4404–8.PubMed
82.
Zurück zum Zitat Basu S, Campbell HM, Dittel BN, Ray A. Purification of specific cell population by fluorescence activated cell sorting (FACS). J Vis Exp. 2010(41): e1546. doi:10.3791/1546. Basu S, Campbell HM, Dittel BN, Ray A. Purification of specific cell population by fluorescence activated cell sorting (FACS). J Vis Exp. 2010(41): e1546. doi:10.​3791/​1546.
83.
Zurück zum Zitat Zaritskaya L, Shurin MR, Sayers TJ, Malyguine AM. New flow cytometric assays for monitoring cell-mediated cytotoxicity. Expert Rev Vaccines. 2010;9(6):601–16.PubMedPubMedCentralCrossRef Zaritskaya L, Shurin MR, Sayers TJ, Malyguine AM. New flow cytometric assays for monitoring cell-mediated cytotoxicity. Expert Rev Vaccines. 2010;9(6):601–16.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Yuan J, Gnjatic S, Li H, Powel S, Gallardo HF, Ritter E, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105(51):20410–5.PubMedPubMedCentralCrossRef Yuan J, Gnjatic S, Li H, Powel S, Gallardo HF, Ritter E, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105(51):20410–5.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Attig S, Price L, Janetzki S, Kalos M, Pride M, McNeil L, et al. A critical assessment for the value of markers to gate-out undesired events in HLA-peptide multimer staining protocols. J Transl Med. 2011;9:108.PubMedPubMedCentralCrossRef Attig S, Price L, Janetzki S, Kalos M, Pride M, McNeil L, et al. A critical assessment for the value of markers to gate-out undesired events in HLA-peptide multimer staining protocols. J Transl Med. 2011;9:108.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat McNeil LK, Price L, Britten CM, Jaimes M, Maecker H, Odunsi K, et al. A harmonized approach to intracellular cytokine staining gating: results from an international multiconsortia proficiency panel conducted by the cancer immunotherapy consortium (CIC/CRI). Cytometry A. 2013;83(8):728–38.PubMedPubMedCentralCrossRef McNeil LK, Price L, Britten CM, Jaimes M, Maecker H, Odunsi K, et al. A harmonized approach to intracellular cytokine staining gating: results from an international multiconsortia proficiency panel conducted by the cancer immunotherapy consortium (CIC/CRI). Cytometry A. 2013;83(8):728–38.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Vales-Gomez M, Reyburn HT, Erskine RA, Lopez-Botet M, Strominger JL. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J. 1999;18(15):4250–60.PubMedPubMedCentralCrossRef Vales-Gomez M, Reyburn HT, Erskine RA, Lopez-Botet M, Strominger JL. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J. 1999;18(15):4250–60.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Kerr KM, Tsao MS, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR, et al. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10(7):985–9.PubMedCrossRef Kerr KM, Tsao MS, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR, et al. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10(7):985–9.PubMedCrossRef
90.
Zurück zum Zitat Gnjatic S, Ritter E, Buchler MW, Giese NA, Brors B, Frei C, et al. Seromic profiling of ovarian and pancreatic cancer. Proc Natl Acad Sci U S A. 2010;107(11):5088–93.PubMedPubMedCentralCrossRef Gnjatic S, Ritter E, Buchler MW, Giese NA, Brors B, Frei C, et al. Seromic profiling of ovarian and pancreatic cancer. Proc Natl Acad Sci U S A. 2010;107(11):5088–93.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Janetzki S, Price L, Schroeder H, Britten CM, Welters MJ, Hoos A. Guidelines for the automated evaluation of Elispot assays. Nat Protoc. 2015;10(7):1098–115.PubMedCrossRef Janetzki S, Price L, Schroeder H, Britten CM, Welters MJ, Hoos A. Guidelines for the automated evaluation of Elispot assays. Nat Protoc. 2015;10(7):1098–115.PubMedCrossRef
94.
Zurück zum Zitat Malyguine AM, Strobl S, Dunham K, Shurin MR, Sayers TJ. ELISPOT assay for monitoring cytotoxic T lymphocytes (CTL) activity in cancer vaccine clinical trials. Cells. 2012;1(2):111–26.PubMedPubMedCentralCrossRef Malyguine AM, Strobl S, Dunham K, Shurin MR, Sayers TJ. ELISPOT assay for monitoring cytotoxic T lymphocytes (CTL) activity in cancer vaccine clinical trials. Cells. 2012;1(2):111–26.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Janetzki S, Panageas KS, Ben-Porat L, Boyer J, Britten CM, Clay TM, et al. Results and harmonization guidelines from two large-scale international Elispot proficiency panels conducted by the cancer vaccine consortium (CVC/SVI). Cancer Immunol Immunother. 2008;57(3):303–15.PubMedPubMedCentralCrossRef Janetzki S, Panageas KS, Ben-Porat L, Boyer J, Britten CM, Clay TM, et al. Results and harmonization guidelines from two large-scale international Elispot proficiency panels conducted by the cancer vaccine consortium (CVC/SVI). Cancer Immunol Immunother. 2008;57(3):303–15.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Butterfield LH, Buffo MJ. Immunologic monitoring of cancer vaccine trials using the ELISPOT assay. Methods Mol Biol. 2014;1102:71–82.PubMedCrossRef Butterfield LH, Buffo MJ. Immunologic monitoring of cancer vaccine trials using the ELISPOT assay. Methods Mol Biol. 2014;1102:71–82.PubMedCrossRef
97.
Zurück zum Zitat Gabrielsson S, Brichard V, Dhellin O, Dorval T, Bonnerot C. IFN-gamma responses in peptide-treated melanoma patients measured by an ELISPOT assay using allogeneic dendritic cells. Anticancer Res. 2004;24(1):171–7.PubMed Gabrielsson S, Brichard V, Dhellin O, Dorval T, Bonnerot C. IFN-gamma responses in peptide-treated melanoma patients measured by an ELISPOT assay using allogeneic dendritic cells. Anticancer Res. 2004;24(1):171–7.PubMed
98.
Zurück zum Zitat Scheibenbogen C, Lee KH, Stevanovic S, Witzens M, Willhauck M, Waldmann V, et al. Analysis of the T cell response to tumor and viral peptide antigens by an IFNgamma-ELISPOT assay. Int J Cancer. 1997;71(6):932–6.PubMedCrossRef Scheibenbogen C, Lee KH, Stevanovic S, Witzens M, Willhauck M, Waldmann V, et al. Analysis of the T cell response to tumor and viral peptide antigens by an IFNgamma-ELISPOT assay. Int J Cancer. 1997;71(6):932–6.PubMedCrossRef
99.
100.
Zurück zum Zitat Robins HS, Ericson NG, Guenthoer J, O'Briant KC, Tewari M, Drescher CW, et al. Digital genomic quantification of tumor-infiltrating lymphocytes. Sci Transl Med. 2013;5(214):214ra169.PubMedPubMedCentralCrossRef Robins HS, Ericson NG, Guenthoer J, O'Briant KC, Tewari M, Drescher CW, et al. Digital genomic quantification of tumor-infiltrating lymphocytes. Sci Transl Med. 2013;5(214):214ra169.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6(238):238ra270.CrossRef Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6(238):238ra270.CrossRef
102.
Zurück zum Zitat Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74.PubMedCrossRef Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74.PubMedCrossRef
103.
Zurück zum Zitat Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125(9):3413–21.PubMedCrossRef Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125(9):3413–21.PubMedCrossRef
104.
Zurück zum Zitat Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–99.PubMedPubMedCentralCrossRef Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–99.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–8.PubMedCrossRef Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–8.PubMedCrossRef
106.
Zurück zum Zitat Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44.PubMedCrossRef Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44.PubMedCrossRef
107.
Zurück zum Zitat Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M, et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol. 2016. doi: 10.1200/JCO.2016.66.6552. Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M, et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol. 2016. doi: 10.​1200/​JCO.​2016.​66.​6552.
108.
Zurück zum Zitat Hoffmann S, Cepok S, Grummel V, Lehmann-Horn K, Hackermuller J, Stadler PF, et al. HLA-DRB1*0401 and HLA-DRB1*0408 are strongly associated with the development of antibodies against interferon-beta therapy in multiple sclerosis. Am J Hum Genet. 2008;83(2):219–27.PubMedPubMedCentralCrossRef Hoffmann S, Cepok S, Grummel V, Lehmann-Horn K, Hackermuller J, Stadler PF, et al. HLA-DRB1*0401 and HLA-DRB1*0408 are strongly associated with the development of antibodies against interferon-beta therapy in multiple sclerosis. Am J Hum Genet. 2008;83(2):219–27.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat International Multiple Sclerosis Genetics C, Wellcome Trust Case Control C, Sawcer S, Hellenthal G, Pirinen M, Spencer CC, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476(7359):214–9.CrossRef International Multiple Sclerosis Genetics C, Wellcome Trust Case Control C, Sawcer S, Hellenthal G, Pirinen M, Spencer CC, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476(7359):214–9.CrossRef
110.
Zurück zum Zitat Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res. 2004;10(15):5094–100.PubMedCrossRef Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res. 2004;10(15):5094–100.PubMedCrossRef
111.
Zurück zum Zitat Chen YY, Wang LB, Zhu HL, Li XY, Zhu YP, Yin YL, et al. Relationship between programmed death-ligand 1 and clinicopathological characteristics in non-small cell lung cancer patients. Chin Med Sci J. 2013;28(3):147–51.PubMedCrossRef Chen YY, Wang LB, Zhu HL, Li XY, Zhu YP, Yin YL, et al. Relationship between programmed death-ligand 1 and clinicopathological characteristics in non-small cell lung cancer patients. Chin Med Sci J. 2013;28(3):147–51.PubMedCrossRef
112.
Zurück zum Zitat Massi D, Brusa D, Merelli B, Falcone C, Xue G, Carobbio A, et al. The status of PD-L1 and tumor-infiltrating immune cells predict resistance and poor prognosis in BRAFi-treated melanoma patients harboring mutant BRAFV600. Ann Oncol. 2015;26(9):1980–7.PubMedCrossRef Massi D, Brusa D, Merelli B, Falcone C, Xue G, Carobbio A, et al. The status of PD-L1 and tumor-infiltrating immune cells predict resistance and poor prognosis in BRAFi-treated melanoma patients harboring mutant BRAFV600. Ann Oncol. 2015;26(9):1980–7.PubMedCrossRef
113.
Zurück zum Zitat Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20(19):5064–74.PubMedPubMedCentralCrossRef Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20(19):5064–74.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.PubMedCrossRef Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.PubMedCrossRef
115.
Zurück zum Zitat Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015;350(6266):1387–90.PubMedCrossRef Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015;350(6266):1387–90.PubMedCrossRef
117.
Zurück zum Zitat Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med. 2013;19(6):747–52.PubMedPubMedCentralCrossRef Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med. 2013;19(6):747–52.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. 2016;22(4):433–8.PubMedCrossRef Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. 2016;22(4):433–8.PubMedCrossRef
119.
Zurück zum Zitat Velcheti V, Schalper KA, Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, et al. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Invest. 2014;94(1):107–16.PubMedCrossRef Velcheti V, Schalper KA, Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, et al. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Invest. 2014;94(1):107–16.PubMedCrossRef
120.
Zurück zum Zitat Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537–44.PubMedPubMedCentralCrossRef Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537–44.PubMedPubMedCentralCrossRef
121.
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(7528):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(7528):568–71.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Yung S, Ledran M, Moreno-Gimeno I, Conesa A, Montaner D, Dopazo J, et al. Large-scale transcriptional profiling and functional assays reveal important roles for Rho-GTPase signalling and SCL during haematopoietic differentiation of human embryonic stem cells. Hum Mol Genet. 2011;20(24):4932–46.PubMedCrossRef Yung S, Ledran M, Moreno-Gimeno I, Conesa A, Montaner D, Dopazo J, et al. Large-scale transcriptional profiling and functional assays reveal important roles for Rho-GTPase signalling and SCL during haematopoietic differentiation of human embryonic stem cells. Hum Mol Genet. 2011;20(24):4932–46.PubMedCrossRef
123.
Zurück zum Zitat Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014;232(2):199–209.PubMedPubMedCentralCrossRef Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014;232(2):199–209.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Shay T, Jojic V, Zuk O, Rothamel K, Puyraimond-Zemmour D, Feng T, et al. Conservation and divergence in the transcriptional programs of the human and mouse immune systems. Proc Natl Acad Sci U S A. 2013;110(8):2946–51.PubMedPubMedCentralCrossRef Shay T, Jojic V, Zuk O, Rothamel K, Puyraimond-Zemmour D, Feng T, et al. Conservation and divergence in the transcriptional programs of the human and mouse immune systems. Proc Natl Acad Sci U S A. 2013;110(8):2946–51.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–35.PubMedCrossRef Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–35.PubMedCrossRef
126.
Zurück zum Zitat Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedCrossRef Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedCrossRef
127.
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(19):1803–13.PubMedCrossRef 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(19):1803–13.PubMedCrossRef
128.
Zurück zum Zitat Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–84.PubMedCrossRef Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–84.PubMedCrossRef
129.
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(4):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(4):320–30.PubMedCrossRef
130.
Zurück zum Zitat Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.PubMedCrossRef Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.PubMedCrossRef
131.
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(3):257–65.PubMedCrossRef 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(3):257–65.PubMedCrossRef
132.
Zurück zum Zitat McDermott DF, Drake CG, Sznol M, Choueiri TK, Powderly JD, Smith DC, et al. Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab. J Clin Oncol. 2015;33(18):2013–20.PubMedCrossRef McDermott DF, Drake CG, Sznol M, Choueiri TK, Powderly JD, Smith DC, et al. Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab. J Clin Oncol. 2015;33(18):2013–20.PubMedCrossRef
133.
Zurück zum Zitat Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33(18):2004–12.PubMedCrossRef Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33(18):2004–12.PubMedCrossRef
134.
Zurück zum Zitat Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–32.PubMedCrossRef Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–32.PubMedCrossRef
135.
Zurück zum Zitat Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–18.PubMedCrossRef Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–18.PubMedCrossRef
136.
Zurück zum Zitat Daud A, Ribas A, Robert C, Hodi FS, Wolchok JD, Joshua AM, et al. Long-term efficacy of pembrolizumab (pembro; MK-3475) in a pooled analysis of 655 patients (pts) with advanced melanoma (MEL) enrolled in KEYNOTE-001. ASCO Meeting Abstr. 2015;33(15_suppl):9005. Daud A, Ribas A, Robert C, Hodi FS, Wolchok JD, Joshua AM, et al. Long-term efficacy of pembrolizumab (pembro; MK-3475) in a pooled analysis of 655 patients (pts) with advanced melanoma (MEL) enrolled in KEYNOTE-001. ASCO Meeting Abstr. 2015;33(15_suppl):9005.
137.
Zurück zum Zitat Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA. 2016;315(15):1600–9.PubMedCrossRef Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA. 2016;315(15):1600–9.PubMedCrossRef
138.
Zurück zum Zitat Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384(9948):1109–17.PubMedCrossRef Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384(9948):1109–17.PubMedCrossRef
139.
Zurück zum Zitat Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, et al. Abstract S1-09: a phase Ib study of pembrolizumab (MK-3475) in patients with advanced triple-negative breast cancer. Cancer Res. 2015;75(9 Supplement):S1–09.CrossRef Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, et al. Abstract S1-09: a phase Ib study of pembrolizumab (MK-3475) in patients with advanced triple-negative breast cancer. Cancer Res. 2015;75(9 Supplement):S1–09.CrossRef
140.
Zurück zum Zitat Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2015;387(10027):1540–50.PubMedCrossRef Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2015;387(10027):1540–50.PubMedCrossRef
141.
Zurück zum Zitat Moskowitz C, Ribrag V, Michot J-M, Martinelli G, Zinzani PL, Gutierrez M, et al. PD-1 Blockade with the monoclonal antibody Pembrolizumab (M-3475) in patients with classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Preliminary results from a phase 1b study [abstract]. Blood. 2014;124:290. abstract. Moskowitz C, Ribrag V, Michot J-M, Martinelli G, Zinzani PL, Gutierrez M, et al. PD-1 Blockade with the monoclonal antibody Pembrolizumab (M-3475) in patients with classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Preliminary results from a phase 1b study [abstract]. Blood. 2014;124:290. abstract.
142.
Zurück zum Zitat Spigel DR, Chaft JE, Gettinger SN, Chao BH, Dirix LY, Schmid P, et al. Clinical activity and safety from a phase II study (FIR) of MPDL3280A (anti-PDL1) in PD-L1-selected patients with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8028. Spigel DR, Chaft JE, Gettinger SN, Chao BH, Dirix LY, Schmid P, et al. Clinical activity and safety from a phase II study (FIR) of MPDL3280A (anti-PDL1) in PD-L1-selected patients with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8028.
143.
Zurück zum Zitat Spira AI, Park K, Mazieres J, Vansteenkiste JF, Rittmeyer A, Ballinger M, et al. Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDL3280A vs docetaxel in 2L/3L NSCLC (POPLAR). ASCO Meeting Abstr. 2015;33(15_suppl):8010. Spira AI, Park K, Mazieres J, Vansteenkiste JF, Rittmeyer A, Ballinger M, et al. Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDL3280A vs docetaxel in 2L/3L NSCLC (POPLAR). ASCO Meeting Abstr. 2015;33(15_suppl):8010.
144.
Zurück zum Zitat Hamid O, Sosman JA, Lawrence DP, Sullivan RJ, Ibrahim N, Kluger HM, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). ASCO Meeting Abstr. 2013;31(15_suppl):9010. Hamid O, Sosman JA, Lawrence DP, Sullivan RJ, Ibrahim N, Kluger HM, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). ASCO Meeting Abstr. 2013;31(15_suppl):9010.
145.
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(7528):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(7528):563–7.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Liu SV, Powderly JD, Camidge DR, Ready N, Heist RS, Hodi FS, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with platinum-based doublet chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8030. Liu SV, Powderly JD, Camidge DR, Ready N, Heist RS, Hodi FS, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with platinum-based doublet chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8030.
147.
Zurück zum Zitat Bendell JC, Powderly JD, Lieu CH, Eckhardt SG, Hurwitz H, Hochster HS, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or FOLFOX in patients (pts) with metastatic colorectal cancer (mCRC). ASCO Meeting Abstr. 2015;33(3_suppl):704. Bendell JC, Powderly JD, Lieu CH, Eckhardt SG, Hurwitz H, Hochster HS, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or FOLFOX in patients (pts) with metastatic colorectal cancer (mCRC). ASCO Meeting Abstr. 2015;33(3_suppl):704.
148.
Zurück zum Zitat Sznol M, McDermott DF, Jones SF, Mier JW, Waterkamp D, Rossi C, et al. Phase Ib evaluation of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) in patients (pts) with metastatic renal cell carcinoma (mRCC). ASCO Meeting Abstr. 2015;33(7_suppl):410. Sznol M, McDermott DF, Jones SF, Mier JW, Waterkamp D, Rossi C, et al. Phase Ib evaluation of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) in patients (pts) with metastatic renal cell carcinoma (mRCC). ASCO Meeting Abstr. 2015;33(7_suppl):410.
149.
Zurück zum Zitat Rizvi NA, Brahmer JR, Ou S-HI, Segal NH, Khleif S, Hwu W-J, et al. Safety and clinical activity of MEDI4736, an anti-programmed cell death-ligand 1 (PD-L1) antibody, in patients with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8032. Rizvi NA, Brahmer JR, Ou S-HI, Segal NH, Khleif S, Hwu W-J, et al. Safety and clinical activity of MEDI4736, an anti-programmed cell death-ligand 1 (PD-L1) antibody, in patients with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):8032.
150.
Zurück zum Zitat Brahmer JR, Rizvi NA, Lutzky J, Khleif S, Blake-Haskins A, Li X, et al. Clinical activity and biomarkers of MEDI4736, an anti-PD-L1 antibody, in patients with NSCLC. ASCO Meeting Abstr. 2014;32(15_suppl):8021. Brahmer JR, Rizvi NA, Lutzky J, Khleif S, Blake-Haskins A, Li X, et al. Clinical activity and biomarkers of MEDI4736, an anti-PD-L1 antibody, in patients with NSCLC. ASCO Meeting Abstr. 2014;32(15_suppl):8021.
151.
Zurück zum Zitat Segal NH, Ou S-HI, Balmanoukian AS, Fury MG, Massarelli E, Brahmer JR, et al. Safety and efficacy of MEDI4736, an anti-PD-L1 antibody, in patients from a squamous cell carcinoma of the head and neck (SCCHN) expansion cohort. ASCO Meeting Abstr. 2015;33(15_suppl):3011. Segal NH, Ou S-HI, Balmanoukian AS, Fury MG, Massarelli E, Brahmer JR, et al. Safety and efficacy of MEDI4736, an anti-PD-L1 antibody, in patients from a squamous cell carcinoma of the head and neck (SCCHN) expansion cohort. ASCO Meeting Abstr. 2015;33(15_suppl):3011.
152.
Zurück zum Zitat Lutzky J, Antonia SJ, Blake-Haskins A, Li X, Robbins PB, Shalabi AM, et al. A phase 1 study of MEDI4736, an anti-PD-L1 antibody, in patients with advanced solid tumors. ASCO Meeting Abstr. 2014;32(15_suppl):3001. Lutzky J, Antonia SJ, Blake-Haskins A, Li X, Robbins PB, Shalabi AM, et al. A phase 1 study of MEDI4736, an anti-PD-L1 antibody, in patients with advanced solid tumors. ASCO Meeting Abstr. 2014;32(15_suppl):3001.
153.
Zurück zum Zitat Creelan BC, Chow LQ, Kim D-W, Kim S-W, Yeh T, Karakunnel JJ, et al. Safety and tolerability results from a phase I study of MEDI4736, a human IgG1 anti-programmed cell death-ligand-1 (PD-L1) antibody, combined with gefitinib in patients (pts) with non-small-cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):3047. Creelan BC, Chow LQ, Kim D-W, Kim S-W, Yeh T, Karakunnel JJ, et al. Safety and tolerability results from a phase I study of MEDI4736, a human IgG1 anti-programmed cell death-ligand-1 (PD-L1) antibody, combined with gefitinib in patients (pts) with non-small-cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(15_suppl):3047.
154.
Zurück zum Zitat Antonia SJ, Goldberg SB, Balmanoukian AS, Sanborn RE, Steele K, Narwal R, et al. Phase Ib study of MEDI4736, a programmed cell death ligand-1 (PD-L1) antibody, in combination with tremelimumab, a cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) antibody, in patients (pts) with advanced NSCLC. ASCO Meeting Abstr. 2015;33(15_suppl):3014. Antonia SJ, Goldberg SB, Balmanoukian AS, Sanborn RE, Steele K, Narwal R, et al. Phase Ib study of MEDI4736, a programmed cell death ligand-1 (PD-L1) antibody, in combination with tremelimumab, a cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) antibody, in patients (pts) with advanced NSCLC. ASCO Meeting Abstr. 2015;33(15_suppl):3014.
155.
Zurück zum Zitat Antonia S, Goldberg SB, Balmanoukian A, Chaft JE, Sanborn RE, Gupta A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17(3):299–308.PubMedCrossRef Antonia S, Goldberg SB, Balmanoukian A, Chaft JE, Sanborn RE, Gupta A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17(3):299–308.PubMedCrossRef
156.
Zurück zum Zitat Paz-Ares L, Horn L, Borghaei H, Spigel DR, Steins M, Ready N, et al. Phase III, randomized trial (CheckMate 057) of nivolumab (NIVO) versus docetaxel (DOC) in advanced non-squamous cell (non-SQ) non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(18_suppl):LBA109. Paz-Ares L, Horn L, Borghaei H, Spigel DR, Steins M, Ready N, et al. Phase III, randomized trial (CheckMate 057) of nivolumab (NIVO) versus docetaxel (DOC) in advanced non-squamous cell (non-SQ) non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2015;33(18_suppl):LBA109.
157.
Zurück zum Zitat Gettinger SN, Hellmann MD, Shepherd FA, Antonia SJ, Brahmer JR, Chow LQM, et al. First-line monotherapy with nivolumab (NIVO; anti-programmed death-1 [PD-1]) in advanced non-small cell lung cancer (NSCLC): safety, efficacy and correlation of outcomes with PD-1 ligand (PD-L1) expression. ASCO Meeting Abstr. 2015;33(15_suppl):8025. Gettinger SN, Hellmann MD, Shepherd FA, Antonia SJ, Brahmer JR, Chow LQM, et al. First-line monotherapy with nivolumab (NIVO; anti-programmed death-1 [PD-1]) in advanced non-small cell lung cancer (NSCLC): safety, efficacy and correlation of outcomes with PD-1 ligand (PD-L1) expression. ASCO Meeting Abstr. 2015;33(15_suppl):8025.
158.
Zurück zum Zitat Antonia SJ, Gettinger SN, Chow LQM, Juergens RA, Borghaei H, Shen Y, et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) and ipilimumab in first-line NSCLC: interim phase I results. ASCO Meeting Abstr. 2014;32(15_suppl):8023. Antonia SJ, Gettinger SN, Chow LQM, Juergens RA, Borghaei H, Shen Y, et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) and ipilimumab in first-line NSCLC: interim phase I results. ASCO Meeting Abstr. 2014;32(15_suppl):8023.
159.
Zurück zum Zitat Rizvi NA, Garon EB, Leighl N, Hellmann MD, Patnaik A, Gandhi L, et al. Optimizing PD-L1 as a biomarker of response with pembrolizumab (pembro; MK-3475) as first-line therapy for PD-L1-positive metastatic non-small cell lung cancer (NSCLC): Updated data from KEYNOTE-001. ASCO Meeting Abstr. 2015;33(15_suppl):8026. Rizvi NA, Garon EB, Leighl N, Hellmann MD, Patnaik A, Gandhi L, et al. Optimizing PD-L1 as a biomarker of response with pembrolizumab (pembro; MK-3475) as first-line therapy for PD-L1-positive metastatic non-small cell lung cancer (NSCLC): Updated data from KEYNOTE-001. ASCO Meeting Abstr. 2015;33(15_suppl):8026.
160.
Zurück zum Zitat Garon EB, Gandhi L, Rizvi N, Hui R, Balmanoukian AS, Patnaik A, et al. Antitumor activity of pembrolizumab (pembro; mk-3475) and correlation with programmed death ligand 1 (pd-l1) expression in a pooled analysis of patients (pts) with advanced non–small cell lung carcinoma (NSCLC). Ann Oncol. 2014;25 suppl 4:LBA43. Garon EB, Gandhi L, Rizvi N, Hui R, Balmanoukian AS, Patnaik A, et al. Antitumor activity of pembrolizumab (pembro; mk-3475) and correlation with programmed death ligand 1 (pd-l1) expression in a pooled analysis of patients (pts) with advanced non–small cell lung carcinoma (NSCLC). Ann Oncol. 2014;25 suppl 4:LBA43.
161.
Zurück zum Zitat Garon EB, Leighl NB, Rizvi NA, Blumenschein GR, Balmanoukian AS, Eder JP, et al. Safety and clinical activity of MK-3475 in previously treated patients (pts) with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2014;32(15_suppl):8020. Garon EB, Leighl NB, Rizvi NA, Blumenschein GR, Balmanoukian AS, Eder JP, et al. Safety and clinical activity of MK-3475 in previously treated patients (pts) with non-small cell lung cancer (NSCLC). ASCO Meeting Abstr. 2014;32(15_suppl):8020.
162.
Zurück zum Zitat Horn L, Spigel DR, Gettinger SN, Antonia SJ, Gordon MS, Herbst RS, et al. Clinical activity, safety and predictive biomarkers of the engineered antibody MPDL3280A (anti-PDL1) in non-small cell lung cancer (NSCLC): update from a phase Ia study. ASCO Meeting Abstr. 2015;33(15_suppl):8029. Horn L, Spigel DR, Gettinger SN, Antonia SJ, Gordon MS, Herbst RS, et al. Clinical activity, safety and predictive biomarkers of the engineered antibody MPDL3280A (anti-PDL1) in non-small cell lung cancer (NSCLC): update from a phase Ia study. ASCO Meeting Abstr. 2015;33(15_suppl):8029.
163.
Zurück zum Zitat Segal NH, Antonia SJ, Brahmer JR, Maio M, Blake-Haskins A, Li X, et al. Preliminary data from a multi-arm expansion study of MEDI4736, an anti-PD-L1 antibody. ASCO Meeting Abstr. 2014;32(15_suppl):3002. Segal NH, Antonia SJ, Brahmer JR, Maio M, Blake-Haskins A, Li X, et al. Preliminary data from a multi-arm expansion study of MEDI4736, an anti-PD-L1 antibody. ASCO Meeting Abstr. 2014;32(15_suppl):3002.
164.
Zurück zum Zitat Loughlin PM, Cooke TG, George WD, Gray AJ, Stott DI, Going JJ. Quantifying tumour-infiltrating lymphocyte subsets: a practical immuno-histochemical method. J Immunol Methods. 2007;321(1-2):32–40.PubMedCrossRef Loughlin PM, Cooke TG, George WD, Gray AJ, Stott DI, Going JJ. Quantifying tumour-infiltrating lymphocyte subsets: a practical immuno-histochemical method. J Immunol Methods. 2007;321(1-2):32–40.PubMedCrossRef
165.
Zurück zum Zitat Carvajal-Hausdorf DE, Schalper KA, Neumeister VM, Rimm DL. Quantitative measurement of cancer tissue biomarkers in the lab and in the clinic. Lab Invest. 2015;95(4):385–96.PubMedPubMedCentralCrossRef Carvajal-Hausdorf DE, Schalper KA, Neumeister VM, Rimm DL. Quantitative measurement of cancer tissue biomarkers in the lab and in the clinic. Lab Invest. 2015;95(4):385–96.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Chen KH, Yuan CT, Tseng LH, Shun CT, Yeh KH. Case report: mismatch repair proficiency and microsatellite stability in gastric cancer may not predict programmed death-1 blockade resistance. J Hematol Oncol. 2016;9(1):29.PubMedPubMedCentralCrossRef Chen KH, Yuan CT, Tseng LH, Shun CT, Yeh KH. Case report: mismatch repair proficiency and microsatellite stability in gastric cancer may not predict programmed death-1 blockade resistance. J Hematol Oncol. 2016;9(1):29.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Shafer-Weaver K, Rosenberg S, Strobl S, Gregory Alvord W, Baseler M, Malyguine A. Application of the granzyme B ELISPOT assay for monitoring cancer vaccine trials. J Immunother. 2006;29(3):328–35.PubMedCrossRef Shafer-Weaver K, Rosenberg S, Strobl S, Gregory Alvord W, Baseler M, Malyguine A. Application of the granzyme B ELISPOT assay for monitoring cancer vaccine trials. J Immunother. 2006;29(3):328–35.PubMedCrossRef
168.
Zurück zum Zitat Barrera L, Rodriguez OA, Morales-Flores R, Garcia-Vicente A, Servín EM, Salinas-Parra F, et al. MINI 02.03 over expression of CD47, decrease of apoptosis and phagocytosis of neutrophils in advanced non-small cell lung cancer patients. J Thoracic Oncol. 2015;10(9_suppl 2):S266. Barrera L, Rodriguez OA, Morales-Flores R, Garcia-Vicente A, Servín EM, Salinas-Parra F, et al. MINI 02.03 over expression of CD47, decrease of apoptosis and phagocytosis of neutrophils in advanced non-small cell lung cancer patients. J Thoracic Oncol. 2015;10(9_suppl 2):S266.
169.
Zurück zum Zitat Streitz M, Miloud T, Kapinsky M, Reed MR, Magari R, Geissler EK, et al. Standardization of whole blood immune phenotype monitoring for clinical trials: panels and methods from the ONE study. Transplant Res. 2013;2(1):17.PubMedPubMedCentralCrossRef Streitz M, Miloud T, Kapinsky M, Reed MR, Magari R, Geissler EK, et al. Standardization of whole blood immune phenotype monitoring for clinical trials: panels and methods from the ONE study. Transplant Res. 2013;2(1):17.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat van Dongen JJ, Lhermitte L, Bottcher S, Almeida J, van der Velden VH, Flores-Montero J, et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia. 2012;26(9):1908–75.PubMedPubMedCentralCrossRef van Dongen JJ, Lhermitte L, Bottcher S, Almeida J, van der Velden VH, Flores-Montero J, et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia. 2012;26(9):1908–75.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Zhao S, Fung-Leung WP, Bittner A, Ngo K, Liu X. Comparison of RNA-Seq and microarray in transcriptome profiling of activated T cells. PLoS One. 2014;9(1):e78644.PubMedPubMedCentralCrossRef Zhao S, Fung-Leung WP, Bittner A, Ngo K, Liu X. Comparison of RNA-Seq and microarray in transcriptome profiling of activated T cells. PLoS One. 2014;9(1):e78644.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Kammula US, Serrano OK. Use of high throughput qPCR screening to rapidly clone low frequency tumour specific T-cells from peripheral blood for adoptive immunotherapy. J Transl Med. 2008;6:60.PubMedPubMedCentralCrossRef Kammula US, Serrano OK. Use of high throughput qPCR screening to rapidly clone low frequency tumour specific T-cells from peripheral blood for adoptive immunotherapy. J Transl Med. 2008;6:60.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Lin SH, He J, Edelman M, Xu T, Gao H, Reuben J, et al. MINI 02.04 sequential assessment of DNA damage response and PD-L1 expression in circulating cells of lung cancer patients during treatment with radiotherapy. J Thoracic Oncol. 2015;10(9_suppl 2):S266–7. Lin SH, He J, Edelman M, Xu T, Gao H, Reuben J, et al. MINI 02.04 sequential assessment of DNA damage response and PD-L1 expression in circulating cells of lung cancer patients during treatment with radiotherapy. J Thoracic Oncol. 2015;10(9_suppl 2):S266–7.
174.
Zurück zum Zitat Adams DL, Martin SS, Alpaugh RK, Charpentier M, Tsai S, Bergan RC, et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc Natl Acad Sci U S A. 2014;111(9):3514–9.PubMedPubMedCentralCrossRef Adams DL, Martin SS, Alpaugh RK, Charpentier M, Tsai S, Bergan RC, et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc Natl Acad Sci U S A. 2014;111(9):3514–9.PubMedPubMedCentralCrossRef
Metadaten
Titel
Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy
verfasst von
Weijie Ma
Barbara M. Gilligan
Jianda Yuan
Tianhong Li
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2016
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-016-0277-y

Weitere Artikel der Ausgabe 1/2016

Journal of Hematology & Oncology 1/2016 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

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