A mass spectrometry analysis was performed using serum from patients receiving checkpoint inhibitors to define baseline protein signatures associated with outcome in metastatic melanoma. Pretreatment serum was obtained from a development set of 119 melanoma patients on a trial of nivolumab with or without a multipeptide vaccine and from patients receiving pembrolizumab, nivolumab, ipilimumab, or both nivolumab and ipilimumab. Spectra were obtained using matrix-assisted laser desorption/ionization time of flight mass spectrometry. These data combined with clinical data identified patients with better or worse outcomes. The test was applied to five independent patient cohorts treated with checkpoint inhibitors and its biology investigated using enrichment analyses. A signature consisting of 209 proteins or peptides was associated with progression-free and overall survival in a multivariate analysis. The test performance across validation cohorts was consistent with the development set results. A pooled analysis, stratified by set, demonstrated a significantly better overall survival for “sensitive” relative to “resistant” patients, HR = 0.15 (95% confidence interval: 0.06–0.40, P < 0.001). The test was also associated with survival in a cohort of ipilimumab-treated patients. Test classification was found to be associated with acute phase reactant, complement, and wound healing pathways. We conclude that a pretreatment signature of proteins, defined by mass spectrometry analysis and machine learning, predicted survival in patients receiving PD-1 blocking antibodies. This signature of proteins was associated with acute phase reactants and elements of wound healing and the complement cascade. This signature merits further study to determine if it identifies patients who would benefit from PD-1 blockade. Cancer Immunol Res; 6(1); 79–86. ©2017 AACR.

Encouraging clinical results in metastatic melanoma with the antibodies nivolumab and pembrolizumab to anti–PD-1 have led to clinically and statistically significant improvements in progression-free and overall survival (OS) compared with alternative first- and second-line therapies (1–3). The promise of these treatments is the induction of durable responses and clinical benefit in a subgroup of around 30% of patients (4). The selection of patients who would benefit from anti–PD-1 therapy based on pretreatment parameters would further clinical understanding of how PD-1 blockade functions and speed development of alternative treatments for the patients not likely to benefit from the use of PD-1 antibodies. Delivering treatments only to those patients likely to benefit would also result in substantial health care savings and decreased morbidity due to elimination of unnecessary toxicity.

Considerable effort has been directed toward establishing the utility of PD-L1 expression on tumor-and/or immune-infiltrating cells measured by immunohistochemistry (IHC), which is currently the best candidate biomarker for selection of patients for anti–PD-1 therapy. Correlations between PD-L1 expression and outcome with PD-1/PD-L1 antibodies have been observed in many studies (5–8), but melanoma patients with negatively stained tumors may still benefit from anti–PD-1 therapy (9). In nonsquamous non–small cell lung cancer, benefit with PD-1 blockade was greater for those with strong PD-L1 staining (5–7). Three tests of PD-L1 expression by IHC are approved by the FDA to guide treatment decisions in bladder and non–small cell lung cancer as well as melanoma, with different assays using different screening thresholds for PD-L1 positivity (10). There is a lack of standardization and universally accepted cutoffs for IHC staining for PD-L1, rendering comparison of biomarker data across trials difficult. PD-L1 expression is a variable, heterogeneous, and dynamic marker inducible by the amounts of IFNγ generated by infiltrating T cells (11). PD-L1 expression thus has limited utility as a predictive marker for the efficacy of PD-1/PD-L1 blockade. A variety of other tumor-associated markers have been found to be associated with resistance to checkpoint inhibition, including infiltration with CD8+PD-1+ T cells (9, 12), altered β-catenin signaling (13, 14), PTEN deletion (15), and a gene signature of IFNγ signaling (16, 17). These markers have yet to be validated in large cohorts of patients.

A serum-based pretreatment test of circulating proteins would not require tissue, and, if found to be associated with a favorable response to PD-1 blocking antibodies would be clinically useful. To develop such a test, sera from a development set of 119 patients with stage IV melanoma collected prior to treatment with the PD-1 antibody nivolumab were analyzed using matrix-assisted laser desorption/ionization—time of flight (MALDI-TOF) mass spectrometry (18, 19). This is a “soft” ionization technique designed to preserve the integrity of large macromolecules. Machine-learning techniques were used to combine the clinical and mass spectral (MS) data to generate a signature of protein peaks associated with outcome after PD-1 blockade (20). We show the results of test development and blinded validation on four independent pretreatment sample sets from patients treated with checkpoint inhibitors, as well as a pooled analysis of patients treated with anti–PD-1 blockade and an analysis of a small cohort of patients treated with a combination of PD-1 and CTLA4 agents. In addition, we show results of a protein set enrichment analysis (PSEA) to elucidate biological pathways associated with test classification.

Sample sets

Six pretreatment serum sample sets were used for test development and validation. Patient characteristics are described in Table 1. For development, we used all available (119) pretreatment serum samples from a clinical study of 133 patients (21, 22) in which the efficacy of nivolumab monotherapy at 1, 3, and 10 mg/kg with or without a peptide vaccine in second- or later line treatment of metastatic melanoma was evaluated. The majority of patients were treated at 3 mg/kg of nivolumab without vaccine, and no differences were observed by dose or with vaccine in this study in any measure of outcome (21). Pretreatment samples and clinical data for validation sets 1 (PD-1 antibodies) and 4 (ipilimumab) were obtained prospectively and consecutively from available patients receiving off-protocol treatment at approved doses collected by the Yale SPORE in Skin Cancer. Samples for validation set 3 (PD-1 antibodies) were collected as part of clinical practice at National Tumor Institute “Fondazione G. Pascale,” Naples. Fifty-two percent were treated with nivolumab or pembrolizumab in second line and 48% in third line. Validation set 2 was collected prospectively at the Massachusetts General Hospital Cancer Center, predominantly from patients treated in first or second lines for advanced disease. Set 5 (ipilimumab + nivolumab) was from patients treated off-protocol at Yale. Available patient characteristics are summarized in Table 1. All samples was collected with appropriate informed consent as part of institutional review board–approved protocols at the respective institutions. Validation samples were run blinded to the clinical data.

Spectral acquisition and processing

Samples were processed using standardized operating procedures described in the supplementary materials. We used the Deep MALDI method of mass spectrometry (18) on a SimulToF mass spectrometer (Virgin Instruments) to generate reproducible mass spectra from small amounts of serum (<2 μL), showing peaks from a higher abundance range than previously possible by exposing the samples to 400,000 MALDI laser “shots” compared with several thousand used in standard applications (23). The spectra were processed to render them comparable between patients and 351 MS peaks were selected for further analysis by assessing their reproducibility and stability. Sample processing and MS analysis methods are described fully in the supplementary materials. Parameters for these procedures were established using only the 119-sample development set, and this fixed procedure was applied to all other sample sets without modification.

Test development

The Diagnostic Cortex platform (20) incorporates machine learning concepts and advances in deep learning (24) and was designed for test development in cases with more attributes than samples, with the goal of minimizing the potential for overfitting and promoting the ability of the developed tests to generalize to unseen datasets. This is achieved by performing many different splits of the development set into training and test sets, obtaining performance estimates only from the test parts, and taking an average over all these results (25). The method is implemented in a semisupervised manner that allows simultaneous refinement of the test and the classes used in its training, revealing the underlying structure in the MS data associated with the chosen measure of outcome of OS. Full details are provided in the supplementary materials (Supplementary Tables S3–S7 and Supplementary Figs. S1 and S2) and in Roder and colleagues (20). The results of a test developed using this platform that was able to stratify patients into those with better and worse outcomes on PD-1 blockade have been presented previously (26).

The parameters and reference data for the final test were generated solely on the development set, and these were then locked. All validations were performed using these fixed analysis procedures.

Protein set enrichment analysis (PSEA)

This analysis applies the gene set enrichment analysis (GSEA) method to protein expression data (27, 28). This method identifies expression differences that are consistent across prespecified groups or sets of features, in our case, proteins. An additional independent reference set of 49 serum samples with matched mass spectrometry data and protein expression data from the panel of 1,129 proteins measured by SomaLogic was used for this analysis. Specific protein sets were created as the intersection of the list of SomaLogic 1,129 panel targets and results of queries for biological functions from GeneOntology, using AmiGO2 tools and UniProt databases (Supplement). The PSEA method associated test classification (sensitive and resistant) with these biological functions via a rank-based correlation of the measured protein expressions with the test classifications of the reference samples.

Statistical analysis

All analyses, except the PSEA, were obtained using SAS9.3 (SAS Institute). The PSEA was carried out using the method described in (27) using Matlab (MathWorks). Survival/progression-free survival (PFS) plots and medians were generated using the Kaplan–Meier method. All P values are two-sided, except for PSEA, which are defined as described by Subramanian and colleagues (27).

A protein signature associated with PFS and OS with PD-1 antibody

The goal was to build a clinically useful test that identified patients receiving anti–PD-1 who would be likely to experience long survival. We constructed a test composed of multiple subtests, each using a clinically different subset of the development sample set, using the classifier development platform described above and in the supplementary materials (Supplementary Tables S3–S6, and Supplementary Figs. S1 and S2). For a sample to be assigned a test classification of sensitive (i.e., long survival), a sample had to classify uniformly into that group for each subtest. This would identify patients with the best outcomes, as the associated sample has to always demonstrate molecular features associated with good outcome whatever subset of samples is used to create the subtest. We generated a binary classifier for each of seven clinically different subsets (see Supplementary Material). Each of these subtests assigns a good or poor prognosis classification to a sample (by comparing this sample with the samples in the development subset of the test). Samples where at least one of the seven subtests indicated poor prognosis were labeled as “resistant.” The seven subtests utilize different numbers of MS peaks (n = 56, 69, 75, 82, 84, 85, and 85), which are partially overlapping and listed in the Supplementary Materials, so that the overall test uses 209 of the 351 analyzed mass spectrometry peak intensities in the classification algorithm.

The test classified 34 (29%) of the patients in the development set as “sensitive” and 85 (71%) as “resistant.” Kaplan–Meier plots for OS and PFS are shown in Fig. 1A and B, respectively. Summary statistics are shown in the development set column of Table 2. Both OS and PFS show significant separation (P = 0.002 and 0.016, respectively) by test classification with substantial effect sizes for each (hazard ratios of 0.37 and 0.55, respectively). The sensitive group had an excellent two-year survival rate with nivolumab treatment of 67%.

The test was performed independently on the whole cohort a second time several months after initial assessment of the signature using the same mass spectrometer and settings to provide an estimate of technical assay reproducibility. Concordance of sample classification was 94% (112/119).

Test classification was independent of gender, age (<65 versus ≥65), PD-L1 expression (PD-L1 testing protocol outlined in supplementary materials), prior ipilimumab therapy, and nivolumab dose/addition of peptide vaccine (p ≥ 0.148), shown in Supplementary Table S1. The classification was found to be significantly associated with serum LDH level (P = 0.006) and baseline tumor size (P < 0.001). However, the proposed test retained its predictive power in a multivariate analysis including these factors (Table 3) for OS and PFS, (P = 0.002 and 0.022, respectively). PD-L1 expression did not appear as a significant predictor of outcome in multivariate analysis, which may be due to the small subset of patients with available measured PD-L1 expression (n = 37).

Test classification was not found to be associated with the occurrence of severe immune adverse events (Supplementary Table S2), although it should be noted that the number of grade 3 to 4 events was low.

The potential biological relevance of the sensitive and resistant groups was investigated using the PSEA approach to assess whether pathways of biologic significance were associated with test classification. These data are summarized in Table 4. Biological processes significantly associated with classification with a false discovery rate <0.25 as assessed by the Benjamini–Hochberg approach (29) included the complement, wound healing, and acute phase reactant pathways. Based on the proteins that are included in the PSEA analysis protein sets (shown in Supplementary Table S8), these processes all appear to be upregulated in the resistant group compared with the sensitive group. Proteins included in these protein sets that were significantly correlated (Mann–Whitney P < 0.05) with the test classification in the reference sample set are listed in Table 5.

Validation

The 209-protein signature was validated in a blinded manner in four independent sample sets from three additional institutions, one for patients treated with ipilimumab and three for patients treated with the PD-1 antibodies nivolumab or pembrolizumab. The results for OS are shown in Fig. 1C–F, and performance estimates of the test are summarized in the respective columns of Table 2. Given the small cohort sizes and limited follow up in some cohorts, for anti–PD-1 antibodies nivolumab and pembrolizumab (validation sets 1–3) the results are consistent with the development data for OS. A pooled analysis of these validation sets, stratified by set, demonstrated a significantly better OS for the patients classified as “sensitive” relative to “resistant,” HR = 0.15 [95% confidence interval (CI): 0.06−0.40, P < 0.001]. PFS data were not available for validation set 1, and although the numerical estimate of hazard ratio between sensitive and resistant groups was the same as the development set for validation set 3 and somewhat larger for validation set 2, the set-stratified pooled analysis for the two sets with available PFS data did not show significantly better PFS for the sensitive group [HR = 0.63 (95% CI: 0.29−1.36), P = 0.239]. The ipilimumab-treated validation set 4 showed a significant difference in OS between sensitive and resistant groups (HR = 0.40, P = 0.004), even with the lower overall efficacy of this therapy compared with PD-1 or combination blockade.

In validation set 5, shown in Fig. 1G, where patients were treated with the combination of ipilimumab and nivolumab, 2-year survival rates were similar and high in both the sensitive and the resistant group (83% and 63%, respectively), and there was no statistically significant separation of the groups by survival. This raises the issue of whether outcome with combination therapy is not associated with the signature, but it requires further analysis with larger numbers of patients to draw any firm conclusions.

Definition of tumor biomarkers that predict response to and benefit from PD-1/PD-L1 blocking antibodies (9–17) would be clinically valuable. Following assays of the tumor microenvironment, the emerging consensus is that patients with T-cell-infiltrated tumors within an inflammatory milieu are most likely to respond to PD-1 blockade (12, 17). In contrast, little attention has been paid to developing serum or blood-based markers due to the concern that such markers may not reflect the immune events unfolding within the tumor. A biomarker strategy focused on the tumor suffers from the heterogeneity of gene expression within tumors, variability from one metastatic lesion to another, access to sufficient tumor for analysis, as well as the context-dependent expression and variations of markers like PD-L1 over time (10). A serum marker would have ease of detection using one blood draw, might be independent of inter- and intratumor variation, and could assess aspects of the host that might not be apparent from the tumor microenvironment. Several serum markers have been suggested to be associated with outcome with checkpoint inhibitors, including soluble PD-L1, and angiopoetin-2 (30–32).

We developed a pretreatment serum test utilizing mass spectrometry to identify a group of serum proteins that distinguish metastatic melanoma patients that went on to have prolonged survival after receiving anti–PD-1 antibodies. The test was validated in a blinded manner in three independent sets of patients treated with PD-1 checkpoint inhibitors. The test measures the relative abundance of 209 circulating proteins or protein fragments via mass spectrometry with MALDI-TOF, and generates a test classification, designated either “sensitive” or “resistant,” by using machine learning. The test is reproducible, and its ability to identify patients likely to have prolonged survival when treated with different PD-1 antibodies was consistent across multiple sets of sera from different institutions and validated in a pooled analysis. The test appears to differentiate a group of patients that have a plateau of survival over 50% from those with less than 20% survival at 3 years. Long-term follow-up of a cohort of nivolumab-treated patients indicated that there was a plateau of the survival curve after 3 years, suggesting that some of those alive at that time point may be cured of melanoma (4).

The serum test described herein might identify patients expressing the “sensitive” serum classification that have long OS with PD-1 blockade alone or with the addition of ipilimumab to nivolumab (33). Although the test was developed in a manner that was agnostic to biological and immunological mechanism, it is important to investigate what differentiates the sensitive and resistant groups from a mechanistic point of view. The PSEA analysis indicated that the serum of PD-1 resistant patients is characterized by acute phase, complement, and wound healing molecules. Inflammatory proteins and acute phase reactants, including CRP, are indicators of poor prognosis in many cancers (34), including melanoma (35–37), and with melanoma patients who received nivolumab (38). Here, we found that complement and wound healing pathways are associated with poor outcomes in patients treated with checkpoint inhibition. In support of these observations, a study established that T cells express C3a and C5a receptors that interact with the IL10 pathway upon ligand binding and suppress tumor specific CD8+ T-cell function (39). An independent study showed that binding of the C3a receptor inhibited neutrophil and CD4+ T-helper activity (40), and C1q has also been shown to act as an immune inhibitor without promoting activation of complement (41). These data indicate that complement activation, or the presence of elements of the complement pathway, may inhibit the efficacy of adaptive antitumor immunity, with a putative role in stimulation of myeloid-derived suppressor cells (41, 42). The role of wound healing in anti–PD-1 resistance was also demonstrated as part of a transcriptomic primary resistance signature, IPRES, and the recruitment of T-regulatory cells in patients with active wound healing may explain the deceased efficacy of checkpoint inhibitors in such patients (17, 43). The current serum test may help in the development of treatments overcoming primary anti–PD-1 resistance by adding inhibition of complement activation, suppression of components of wound healing or down modulation of acute phase pathways by blocking IL6 and IL1 signaling. Further experiments to demonstrate a role for these molecules in antitumor immunity are ongoing.

There are limitations to our data. Our validation sets are relatively small, have limited follow up, and some data such as PD-L1 expression status is only available for a subset of the development cohort. Further validation in much larger retrospective sets with long-term follow-up is needed and is in progress. Although we did not observe association of PD-L1 expression status and test classification in this study, it would be of interest to reassess this in a larger cohort of patients to investigate whether a combination of both markers could provide better stratification of patient outcomes. Before use in clinical practice for patient selection, the utility of this test would have to be shown in prospective randomized studies to distinguish whether the signature represents a prognostic rather than a predictive biomarker. It would also be important to observe changes in the levels of complement and wound healing activation at baseline and over time by a comparison of tumor tissue transcriptome and serum analysis. The availability of a validated serum assay to predict outcome with PD-1 blockade could provide guidance in selecting metastatic melanoma patients for immunotherapy, and for further understanding the mechanisms of sensitivity to, and resistance to, checkpoint blockade.

J.S. Weber has ownership interest (including patents) in Altor, Biond, and Cytomx and is a consultant/advisory board member for Bristol-Myers Squibb, Merck, WindMIL, Novartis, Glaxo Smith Kline, Genentech, Astra Zeneca, EMD Serono, Incyte, Dai Ichi, and Celldex. M. Sznol is a consultant/advisory board member for Biodesix. R.J. Sullivan is a consultant/advisory board member for Biodesix, Merck, Novartis, Amgen, and Takeda. H.M. Kluger reports receiving a commercial research grant from Merck and is a consultant/advisory board member for Biodesix. P.A. Ascierto reports receiving commercial research grants from Bristol-Myers Squibb, Roche-Genentech, and Array; is a consultant/advisory board member for Bristol-Myers Squibb, Roche-Genentech, Genmab, Medimmune, MSD, Array, Novartis, Amgen, Merck Serono, Pierre Fabre, Incyte, and NewLink Genetics. C. Oliveira, H. Roder, J. Roder, J. Grigorieva, S.G. Asmellash, and K. Meyer own stock options and patents with Biodesix. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J.S. Weber, H.M. Kluger, H. Roder

Development of methodology: C. Oliveira, K. Meyer, S.G. Asmellash, J. Roder, H. Roder

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.S. Weber, M. Sznol, R.J. Sullivan, G. Boland, H.M. Kluger, R. Halaban, P.A. Ascierto, M. Capone, S.G. Asmellash, H. Roder

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.S. Weber, H.M. Kluger, P.A. Ascierto, C. Oliveira, K. Meyer, J. Grigorieva, J. Roder, H. Roder

Writing, review, and/or revision of the manuscript: J.S. Weber, M. Sznol, R.J. Sullivan, H.M. Kluger, R. Halaban, P.A. Ascierto, C. Oliveira, J. Grigorieva, S.G. Asmellash, J. Roder, H. Roder

Study supervision: J.S. Weber

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
Nivolumab
in previously untreated melanoma without BRAF mutation
.
N Eng J Med
2015
;
372
:
320
30
.
2.
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
:
908
18
.
3.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Eng J Med
2015
;
372
:
2521
32
.
4.
Hodi
FS
,
Kluger
H
,
Sznol
M
,
Carvajal
R
,
Lawrence
D
,
Atkins
M
, et al
Durable, long-term survival in previously treated patients with advanced melanoma (MEL) who received nivolumab (NIVO) monotherapy in a phase I trial.
Cancer Res
2016
;
76
:
76
(
suppl
).
5.
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 Eng J Med
2015
;
373
:
1627
39
.
6.
Brahmer
J
,
Reckamp
KL
,
Baas
P
,
Crinò
L
,
Eberhardt
WEE
,
Poddubskaya
E
, et al
Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer
.
N Eng J Med
2015
;
373
:
123
35
.
7.
Herbst
RS
,
Baas
P
,
Kim
DW
,
Felip
E
,
Pérez-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
2016
;
387
:
1540
50
.
8.
Herbst
RS
,
Soria
JC
,
Kowanetz
M
,
Fine
GD
,
Hamid
O
,
Gordon
MS
, et al
Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients
.
Nature
2014
;
515
:
563
7
.
9.
Daud
AI
,
Wolchok
JD
,
Robert
C
,
Hwu
W-J
,
Weber
JS
,
Ribas
A
, et al
Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma
.
J Clin Oncol
2016
;
34
:
4102
09
.
10.
Ratcliffe MJ
SA
,
Midha
A
,
Barker
C
,
Barker
C
,
Scorer
P
,
Walker
J
, et al
A comparative study of PD-L1 diagnostic assays and the classification of patients as PD-L1 positive and PD-L1 negative
.
Presented at the AACR Annual Meeting
,
New Orleans, LA
, 
April 16–20, 2016
.
11.
Taube
JM
,
Anders
RA
,
Young
GD
,
Xu
H
,
Sharma
R
,
McMiller
TL
, et al
Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape
.
Sci Transl Med
2012
;
4
:
127ra137
.
12.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
13.
Spranger
S
,
Bao
R
, 
Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
14.
Sweis
RF
,
Spranger
S
,
Bao
R
,
Paner
GP
,
Stadler
WM
,
Steinberg
G
, et al
Molecular drivers of the non-T-cell-inflamed tumor microenvironment in urothelial bladder cancer
.
Cancer Immunol Res
2016
;
4
:
563
8
.
15.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
88
.
16.
Gao
J
,
Shi
LZ
,
Zhao
H
,
Chen
J
,
Xiong
L
,
He
Q
, et al
Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy
.
Cell
2016
;
167
:
397
404.e9
.
17.
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
:
35
44
.
18.
Duncan
MW
,
Asmellash
S
,
Allen
J
, et al
Extending
the information content of the MALDI analysis of biological fluids (Deep MALDI)
.
Presented at the ASMS
,
Minneapolis, MN
, 
June 9–13, 2013
.
19.
Roder
H
,
Asmellash
S
,
Allen
J
,
Tsypin
M
. 
inventors; Biodesix Inc, assignee. Deep-MALDI TOF mass spectrometry of complex biological samples, e.g., serum, and uses thereof
.
United States patent US
2016
;
9
:
279,798
.
20.
Roder
H
,
Roder
J
inventors; Biodesix Inc, assignee
. 
Classification generation method using combination of mini-classifiers with regularization and uses thereof
.
United States patent US
2016
;
9
,
477,906
.
21.
Weber
JS
,
Kudchadkar
RR
,
Yu
B
,
Gallenstein
D
,
Horak
CE
,
Inzunza
HD
, et al
Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma
.
J Clin Oncol
2013
;
31
:
4311
8
.
22.
Weber
J
,
Gibney
G
,
Kudchadkar
R
,
Yu
B
,
Cheng
P
,
Martinez
AJ
, et al
Phase I/II study of metastatic melanoma patients treated with nivolumab who had progressed after ipilimumab
.
Cancer Immunol Res
2016
;
4
:
345
53
.
23.
Singal
N
,
Kumar
M
,
Kanaujia
PK
,
Virdi
JS
. 
MALDI-TOF mass spectrometry: an emerging technology for microbial identification and diagnosis
.
Front. Microbiol
2015
;
6
:
791
.
24.
Goodfellow
I
,
Bengio
Y
,
Courville
A
. 
Deep Learning
,
Cambridge MA
;
MIT Press
2017
.
25.
Breimann
L
. 
Bagging predictors
.
Machine Learning
1996
;
24
:
123
40
.
26.
Weber
J
,
Martinez
Roder H
,
Roder
J
,
Meyer
K
,
Asmellash
S
, et al
Pre-treatment patient selection for nivolumab benefit based on serum mass spectra
.
Presented at SITC, National Harbor
,
MD
, 
2015; Nov 6–8
.
27.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci USA
2005
;
102
:
15545
50
.
28.
Lavallee-Adam
M
,
Rauniyar
N
,
McClatchy D
B
,
Yates
JR
 3rd
. 
PSEA—Quant: a protein set enrichment analysis on label-free and label-based protein quantification data
.
J Proteome Res
2014
;
13
:
5496
509
.
29.
Benjamini
Y
,
Hochberg
Y
. 
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J R Statist Soc B
1995
;
57
:
289
300
.
30.
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 Eng J Med
. 
2015
;
372
:
2018
28
.
31.
Zhou
J
,
Mahoney
KM
,
Giobbie-Hurder
A
,
Zhao
F
,
Lee
S
,
Liao
X
, et al
Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade
.
Cancer Immunol Res
. 
2017
;
5
:
480
92
.
32.
Wu
X
,
Giobbie-Hurder
A
,
Liao
X
,
Connelly
C
,
Connolly
EM
,
Li
J
, et al
Angiopoietin-2 as a biomarker and target for immune checkpoint therapy
.
Cancer Immunol Res
2017
;
5
:
17
28
.
33.
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
:
23
34
.
34.
Pierce
BL
,
Ballard-Barbash
R
,
Bernstein
L
,
Baumgartner
RN
,
Neuhouser
ML
,
Wener
MH
, et al
Elevated biomarkers of inflammation are associated with reduced survival among breast cancer patients
.
J Clin Oncol
2009
;
27
:
3437
44
.
35.
Mian
S
,
Ugurel
S
,
Parkinson
E
,
Schlenzka
I
,
Dryden
I
,
Lancashire
L
, et al
Serum proteomic fingerprinting discriminates between clinical stages and predicts disease progression in melanoma patients
.
J Clin Oncol
2005
;
23
:
5088
93
.
36.
Findeisen
P
,
Zapatka
M
,
Peccerella
T
,
Matzk
H
,
Neumaier
M
,
Schadendorf
D
, et al
Serum amyloid A as a prognostic marker in melanoma identified by proteomic profiling
.
J Clin Oncol
2009
;
27
:
2199
208
.
37.
Fang
S
,
Wang
Y
,
Sui
D
,
Liu
H
,
Liu
H
,
Ross
MI
,
Gershenwald
JE
, et al
C-reactive protein as a marker of melanoma progression
.
J Clin Oncol
2015
;
33
:
1389
96
.
38.
Nakamura
Y
,
Kitano
S
,
Takahashi
A
,
Tsutsumida
A1
,
Namikawa
K
,
Tanese
K
, et al
Nivolumab for advanced melanoma: pretreatment prognostic factors and early outcome markers during therapy
.
Oncotarget
2016
;
7
:
77404
15
.
39.
Wang
Y
,
Sun
SN
,
Liu
Q
,
Yu
YY
,
Guo
J
,
Wang
K
, et al
Autocrine complement inhibits IL10-dependent T-cell-mediated antitumor immunity to promote tumor progression
.
Cancer Discov
2016
;
6
:
1022
35
.
40.
Nabizadeh
JA
,
Manthey
HD
,
Steyn
FJ
,
Chen
W
,
Widiapradja
A
,
Md Akhir
FN
, et al
The complement C3a receptor contributes to melanoma tumorigenesis by inhibiting neutrophil and CD4+ T cell responses
.
J Immunol
2016
;
196
:
4783
92
.
41.
Bulla
R
,
Tripodo
C
,
Rami
D
,
Ling
GS
,
Agostinis
C
,
Guarnotta
C
, et al
C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation
.
Nat Commun
2016
;
7
:
10346
.
42.
Markiewski
MM
,
DeAngelis
RA
,
Benencia
F
,
Ricklin-Lichtsteiner
SK
,
Koutoulaki
A
,
Gerard
C
, et al
Modulation of the antitumor immune response by complement
.
Nat Immunol
2008
;
9
:
1225
35
.
43.
Antonio
N
,
Bønnelykke-Behrndtz
ML
,
Ward
LC
,
Collin
J
,
Christensen
IJ
,
Steiniche
T
, et al
The wound inflammatory response exacerbates growth of pre-neoplastic cells and progression to cancer
.
EMBO J
2015
;
34
:
2219
36
.