Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Models predicting tumor neoantigen presentation and T cell receptor recognition help to identify responders to immune-checkpoint inhibitors. Radiomics harbors the potential to evaluate biomarkers for immune-checkpoint blockade without the need to obtain t + 3723 word(s) 3723 2020-04-22 05:00:22 |
2 format correct -5 word(s) 3718 2020-05-13 06:07:08 | |
3 format correct -28 word(s) 3690 2020-10-29 04:18:33 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Huemer, F.; Leisch, M.; Geisberger, R.; Melchardt, T.; Rinnerthaler, G.; Zaborsky, N.; Greil, R. Predicting Response to Immune-Checkpoint Inhibition. Encyclopedia. Available online: https://encyclopedia.pub/entry/682 (accessed on 29 March 2024).
Huemer F, Leisch M, Geisberger R, Melchardt T, Rinnerthaler G, Zaborsky N, et al. Predicting Response to Immune-Checkpoint Inhibition. Encyclopedia. Available at: https://encyclopedia.pub/entry/682. Accessed March 29, 2024.
Huemer, Florian, Michael Leisch, Roland Geisberger, Thomas Melchardt, Gabriel Rinnerthaler, Nadja Zaborsky, Richard Greil. "Predicting Response to Immune-Checkpoint Inhibition" Encyclopedia, https://encyclopedia.pub/entry/682 (accessed March 29, 2024).
Huemer, F., Leisch, M., Geisberger, R., Melchardt, T., Rinnerthaler, G., Zaborsky, N., & Greil, R. (2020, April 27). Predicting Response to Immune-Checkpoint Inhibition. In Encyclopedia. https://encyclopedia.pub/entry/682
Huemer, Florian, et al. "Predicting Response to Immune-Checkpoint Inhibition." Encyclopedia. Web. 27 April, 2020.
Predicting Response to Immune-Checkpoint Inhibition
Edit

The therapeutic concept of unleashing a pre-existing immune response against the tumor by the application of immune-checkpoint inhibitors (ICI) has resulted in long-term survival in advanced cancer patient subgroups. Established biomarkers such as programmed death ligand 1 (PD-L1) and tumor mutational burden (TMB) help to select patients who will most likely benefit from ICI, however, biomarker negativity does not exclude responses. Investigating alterations in the antigen presenting pathway as well as radiomics have the potential to determine tumor immunogenicity and response to ICI. A plethora of steps is crucial for proper tumor neoantigen presentation and T cell recognition. Alterations in the antigen presenting pathway give rise to resistance mechanisms that in turn abolish the effect of ICI. Tumor neoantigen prediction models have been shown to identify cancer patients who benefit most from immune-checkpoint blockade, however, a high false positive rate is a drawback of these models. The predictive value of already established biomarkers is considerably heterogeneous across various malignancies and intratumoral and intrapatient heterogeneity complicate tumor tissue-based biomarker assessment. Radiomics offers the opportunity to evaluate biomarkers based on imaging studies without the necessity to perform tumor tissue biopsies. Several radiomics studies have shown to predict clinical outcome with ICI. Radiomics might also help to identify patients who are at risk for hyperprogressive disease upon initiation of anti-PD-1/anti-PD-L1 therapy and patients who are at risk for high grade immune-related adverse events.

radiomics tumor neoantigen MHC PD-L1 immune-checkpoint T cell receptor repertoire beta 2 microglobulin loss of heterozygosity HLA

1. Alterations in the Antigen Presenting Pathway

The cancer immunity cycle highlights a cascade of steps which are necessitated to produce anti-tumor responses by the immune system [1]. However, a magnitude of escape mechanisms prevent tumor neoantigen recognition and in turn abolish the effect of immune-checkpoint inhibitors (ICI). These escape mechanisms are found at the DNA level (e.g., loss of neoantigens due to chromosomal instability), at the RNA level (e.g., decreased neoantigen expression due to promoter hypermethylation) as well as at the protein level (e.g., gene mutations affecting HLA heterozygosity) [2]. Currently available and/or already established predictive markers for ICI such as programmed death ligand 1 (PD-L1) [3][4][5][6] and tumor mutational burden (TMB) [7] only depict the tip of the iceberg of the cancer immunity cycle. Mutant tumor peptides have to be intracellularly processed into nine to eleven amino acid peptides, which must fit and be presented in the groove of one of the patients’ surface major histocompatibility complex (MHC) I molecules [8]. Aspects of the MHC I processing and presentation pathway in order to predict tumor neoantigens, binding affinity of these tumor neoantigens to MHC I, as well as the T cell receptor (TCR) repertoire have come into the focus of immune-checkpoint blocking strategies. Despite a magnitude of evolving biomarkers for ICI and greatest interest in the gut microbiome [9], antibiotic treatment status [10][11] and T cell exhaustion markers [12], within this subsection we review the literature about tumor neoantigen presentation and prediction with regard to the application of ICI for cancer treatment.

A high TMB has been shown to be a positive predictive marker for clinical outcome with ICI across various tumor entities [13][14][15]. A higher tumor neoantigen burden is associated with improved clinical outcome in advanced non-small cell lung cancer (NSCLC) [15] and advanced melanoma [16] patients undergoing immune-checkpoint blockade and shows a strong correlation with TMB. However, mounting evidence suggests that especially patients with a high clonal neoantigen burden and a low intratumoral neoantigen heterogeneity benefit from ICI [17].

Among 77,803 identified tumor neoantigens, Rizvi et al. only found 28 (0.04%) in more than one melanoma patient [[15]. Comparable findings (99% unique neoantigens) were reported among gastrointestinal tumors [18]. These data corroborate that tumor neoantigens appear to be private events. Neoantigen binding to MHC I is the most selective step leading to peptide presentation. Only 3–4% of predicted tumor neoantigens turn out to be MHC I binders and in turn form neoepitopes [19][20]. Bjerregaard et al. investigated natural T cell responses to predicted tumor neoepitopes. Among 1948 predicted neopeptide-MHC I combinations from 13 publications, the vast majority showed a strong binding affinity to MHC I. However, only 53 neoepitopes (3%) were able to elicit T cell responses [21].

Tumor neoantigen prediction models (as summarized in Table 1) could be of special interest for the application of ICI and key questions to be answered by these models are: which mutated proteins are processed into eight to eleven amino acid peptides by the proteasome, and are transferred into the endoplasmatic reticulum by the transporter associated with antigen processing (TAP), and are loaded onto one out of six MHC I molecules in the individual patient (about 12,000 human leukocyte antigen (HLA) alleles identified in the human population [22]), and are shuttled to the cell surface by chaperone proteins in order to be recognized by cytotoxic T-lymphocytes [23].

Table 1. Overview of (tumor) neoantigen prediction models.

Reference

Publication Date

Author

[24]

1998

Mamitsuka et al.

[25]

2002

Dönnes et al.

[26]

2003

Nielsen et al.

[27]

2005

Larsen et al.

[28]

2006

Antes et al.

[29]

2007

Nielsen et al.

[30]

2008

Lundegaard et al.

[31]

2009

Hoof et al.

[32]

2009

Zhang et al.

[33]

2009

Kim et al.

[34]

2011

Lundegaard et al.

[35]

2013

Calis et al.

[30]

2014

Yadav et al.

[36]

2015

Pedersen et al.

[37]

2016

Andreatta et al.

[38]

2016

Nielsen et al.

[39]

2016

Kalaora et al.

[40]

2017

Jurtz et al.

[41]

2017

McGranahan et al.

[42]

2017

Luksza et al.

[43]

2018

O’Donnell et al.

[44]

2018

Kim et al.

Each of the aforementioned steps is crucial for proper tumor neoantigen presentation. Down-regulation of TAP1 (e.g., by promoter methylation) is associated with a lower infiltration of tumor infiltrating lymphocytes (TILs) and with an inferior clinical outcome in early colorectal cancer (CRC) [[45] and genetic variants of TAP are associated with the development of high-grade cervical neoplasia [46]. Lower expression of HLA class I genes as well as of beta-2 microglobulin (β2m) are immune escape mechanisms in NSCLC [17][47][48] and melanoma [49] patients undergoing immune-checkpoint blockade. HLA class I loss has been shown to prevent continuous T cell recognition in a human melanoma model [50]. HLA-A down-regulation is mediated e.g., by the RNA-binding protein MEX3B [51], by loss of function mutations in the genes encoding the interferon-receptor associated Janus kinase 1 (JAK1) or Janus kinase 2 (JAK2) [52][53][54] or by truncating mutations in the gene encoding beta 2 microglobulin (β2m) [52]. A major impact of HLA class I genotype on clinical outcome with ICI has been corroborated by Chowell et al. HLA-I homozygosity in at least one locus was associated with an inferior survival in two independent cancer cohorts undergoing immune-checkpoint blockade and proved as an independent predictor of survival in multivariate analysis. The combined effect of HLA class I genotype and TMB on survival was greater than the effect of TMB alone [55]. In a similar approach, Goodman et al. reported a better discrimination of survival among TMB high cancer patients undergoing immune checkpoint blockade by considering the MHC I genotype [56]. Prediction models such as the Loss of Heterozygosity in Human Leukocyte Antigen (LOHHLA) bioinformatics tool enable estimation of allele-specific HLA loss from sequencing data and improve neoantigen prediction accuracy [41]. Hopkins et al. examined the role of the peripheral TCR repertoire in immunotherapy treated pancreatic adenocarcinoma. A low baseline clonality as well as a high number of expanded clones following treatment with cytotoxic T-lymphocyte protein 4 (CTLA-4) targeting ipilimumab was associated with a statistically significantly longer survival. The latter results were not reproducible with programmed cell death protein 1 (PD-1) targeting therapy [57]. Comparable findings concerning TCR repertoire dynamics [58] and clinical outcome [58][59] with anti-CTLA-4 and anti-PD-1 targeting therapy were reported in advanced melanoma patients [58]. Despite the limited number of patients included in the aforementioned retrospective analyses, the opposite impact of baseline TCR clonality on clinical outcome with anti-CTLA-4 and anti-PD-1 targeting therapy is hypothesis generating and suggests sequential immunotherapy strategies of anti-CTLA-4 followed by anti-PD-1 targeting therapy.

A high false positive rate remains a major drawback of tumor neoantigen prediction algorithms. MHC class I binding affinity (calculated as the wild-type peptide binding affinity relative to the mutant peptide binding affinity) was demonstrated to be a major determinant of cancer peptide immunogenicity and outperformed TMB as well as neoantigen burden for clinical outcome in melanoma and NSCLC patients undergoing immune-checkpoint blockade [60]. In an integrative approach, Kalaora et al. combined whole-exome and RNA sequencing with MHC-peptidomics (analysis of peptide binding to MHC I by liquid chromatography and tandem mass spectrometry) and the neoantigen prediction tool NETMHCpan in advanced melanoma patients. In a direct comparison, this prediction tool, which integrates binding affinity data and mass spectrometry data, outperformed other neoantigen prediction alogorithms [39]. The latter approach highlights the advantage of combining bioinformatic neoantigen prediction with MHC-peptidomics in order to reduce the rate of false positive neoepitopes, especially in cases of rare HLA allotypes [20][61].

However, peptides with a predicted high MHC I binding affinity are not necessarily immunogenic. In neoepitope prediction strategies, attempts such as the integration of information concerning the hydrophobicity of the TCR contact region [44][62], amino acid characteristics [35] or binding differences between wild-type and mutant epitopes [40] yield at increasing the probability to identify clinically relevant neoepitopes [44]. Calis et al. reported two common properties of neopeptide-MHC combinations, which cause differences in T cell recognition: (1) the composition of amino acids in the position 4-6 of the presented peptide as well as (2) the size and absence/presence of aromatic side chains [35]. Neopepsee, a machine-learning-based neoantigen prediction program, integrates nine immunogenicity features including the aforementioned features and was able to determine immunogenic neoantigens in melanoma and chronic lymphocytic leukemia (CLL). Furthermore, the presence of immunogenic neoantigens determined by Neopepsee was associated with a better prognosis in patients with gastric cancer [40]. Luksza et al. combined estimations of the probability that a neoantigen will be presented on MHC I and the probability that presented neoantigens will be recognized by the TCR repertoire based on tumor clonality, MHC I binding affinity and microbial epitope homology. This model was applied to two melanoma cohorts and one NSCLC cohort undergoing anti-CTLA-4 and anti-PD-1 targeting therapy, respectively, and predicted survival in each cohort [37]. Snyder et al. developed a bioinformatic pipeline incorporating MHC class I binding probability, TCR binding probability, patient specific HLA genotype and epitope-homology analysis in order to identify putative neoepitopes associated with clinical outcome in advanced melanoma patients undergoing anti-CTLA-4 targeting therapy. Among predicted neoantigens, conserved stretches of amino acids were identified that were shared by patients with clinical benefit exceeding six months. These neoepitope signatures were significantly associated with survival in the discovery as well as in the validation set [63]. Published studies evaluating the antigen presenting pathway and TCR repertoire by artificial intelligence and the impact on clinical outcome in patients undergoing immune-checkpoint blockade are summarized in Table 2.

Table 2. Impact of the antigen presenting pathway and T cell receptor (TCR) repertoire on clinical outcome with immune-checkpoint inhibitors (ICI).

Reference

Author

Tumor Entity

Findings

[17]

McGranahan et al.

NSCLC, melanoma

↑ PFS/OS with high clonal neoantigen burden + low intratumoral neoantigen heterogeneity

[47]

Gettinger et al.

NSCLC

β2m loss drives resistance to ICI

[48]

Sade-Feldman et al.

melanoma

β2m LOH drives resistance to ICI

[55]

Chowell et al.

solid tumors

↑ OS with maximal heterozygosity at HLA-I loci

[56]

Goodman et al.

solid tumors

↑ ORR/PFS/OS prediction by MHC I genotype analysis among TMBhigh tumors

[57]

Hopkins et al.

pancreatic ductal adenocarcinoma

↑ OS with low baseline TCR clonality before anti-CTLA-4 Tx

↑ OS with higher number of expanded TCR clones following anti-CTLA-4 Tx

[59]

Hogan et al.

melanoma

↑ ORR/PFS with low baseline TCR clonality in anti-CTLA-4 treated patients

↑ ORR/PFS with high baseline TCR clonality in anti-PD-1 treated patients

[60]

Ghorani et al.

NSCLC, melanoma

↑ PFS/OS prediction by assessment of differential binding affinity of mutated peptides for MHC I compared to TMB or tumor neoantigen burden

[42]

Luksza et al.

NSCLC, melanoma

OS discrimination based on neoantigen MHC I binding affinity and T cell recognition

[63]

Snyder et al.

melanoma

OS prediction based on neoantigen
MHC I binding probability, TCR binding probability, HLA genotype and epitope-homology analysis

PFS: progression-free survival; OS: overall survival; MHC: major histocompatibility complex; TCR: T cell receptor; HLA: human leukocyte antigen; ORR: overall response rate; NSCLC: non-small cell lung cancer; β2m: beta-2 microglobulin; ICI: immune-checkpoint inhibitor; LOH: loss of heterzygosity; TMB: tumor mutational burden; CTLA-4: cytotoxic T-lymphocyte protein 4; Tx: therapy; PD-1: programmed cell death protein 1;.

A plethora of previous studies have focused on individual factors affecting the success of immune-checkpoint blockade in immuno-oncology. However, a comprehensive analysis incorporating multiple factors is of utmost importance. Apart from the antigen presenting pathway, future models predicting clinical outcome with ICI necessitate the integration of additional factors affecting the tumor-host interaction such as PD-L1 expression, gut microbiota composition, patient germline genetics, immune microenvironment composition as well as absence/presence of soluble inhibitory molecules as proposed in several cancer immunograms [12][64][65]. For such an approach, DNA sequencing data of the tumor, RNA sequencing data of the microenvironment and germline DNA sequencing will be required. In this regard, Xie et al. developed a multifactorial deep learning model integrating microsatellite instability (MSI-H) burden, somatic copy number alteration (SCNA) burden and modified TMB (mTMB) into four genomic clusters. Data were derived from 8,646 samples of The Cancer Genome Atlas (TCGA) across 29 tumor types. Interestingly, the abovementioned genomic features only showed a weak to moderate correlation, suggesting that each feature has a distinct impact on tumor biology. The authors used TCGA RNA sequencing data to characterize the tumor microenvironment of each genomic cluster by the level of TIL infiltration, expression of immune genes and status of immune pathways. Each cluster was associated with a unique immune landscape. Genomic clusters discriminated patients with different risk for OS in the entire cohort as well as in multiple cancer types. When applying these four genomic clusters to two anti-CTLA-4 treated melanoma cohorts, cluster 4 (MSIhigh, SCNAhigh, mTMBlow) showed the lowest rate of clinical benefit and the shortest OS [66].

However, prospective validation and reproducibility in a real-world setting will be prerequisites for applying such prediction models in clinical practice.

2. Radiomics

In general, the assessment of predictive biomarkers for ICI is frequently limited by the availability of tumor tissue, intralesional as well as interlesional tumor heterogeneity [67] and by expression dynamics during the course of disease [68] and necessitates invasive procedures with relevant periprocedural risks [69][70] in often comorbid cancer patients.

Due to the availability of routinely performed imaging studies and correlations of images with underlying biological processes radiomics may serve a new predictive tool in immuno-oncology in the near future. Apart from non-invasive identification of potential responders to ICI, addressing resistance mechanisms as well as visualization of drug distribution and of the tumor microenvironment are major goals of radiomics in immuno-oncology. Radiomics is based on common imaging modalities such as computed tomography (CT), positron emission tomography (PET) and magnetic resonance tomography (MRT) and necessitates the following steps: image acquisition, identification of the target volumes, segmentation, feature extraction and analysis [71].

2.1. Assessment of Mutation Status by Radiomics

CT-based radiomic features are associated with molecular aberrations [72][73][74][75] in various types of cancer. Yang et al. found a highly statistically significant association between a CT-based radiomic signature and KRAS/NRAS/BRAF mutations in a test cohort of 61 CRC patients (area under curve (AUC): 0.869, p < 0.001) and confirmed the results in a validation cohort [72]. In the light of the recently reported positive predictive value of KRAS mutations for pembrolizumab monotherapy response in the KEYNOTE-042 study [76] such a radiomic approach could be of clinical relevance for treatment decisions in advanced non-squamous NSCLC. Mismatch repair deficient (dMMR) tumors harbor high numbers of mutation-associated neoantigens and are considered sensitive to ICI [77]. The latter finding in turn has led to the tissue/site-agnostic approval of pembrolizumab in dMMR solid tumors by the FDA. Huang et al. demonstrated the feasibility to assess the mismatch repair status by a CT-based radiomic signature in a test cohort of 140 CRC patients (AUC: 0.914, p < 0.001) and confirmed the good discrimination in a validation cohort including 114 CRC patients (AUC: 0.702, p = 0.012) [73]. Due to the low frequency of dMMR solid tumors in advanced stages [77], this radiomic approach will only identify a minority of potential responders to ICI. NSCLC harboring activating EGFR mutations are insensitive to ICI monotherapy [78][79]. Yip et al. showed the potential of quantitative CT imaging to predict the EGFR mutation status in operable NSCLC patients in the perioperative setting (AUC: 0.67) [176]. Comparable findings based on FDG-PET CT imaging were described by Gevaert et al. in stage 1–4 NSCLC patients (AUC: 0.89) [75].

2.2. PD-1/PD-L1 Expression and Heterogeneity Assessed by Radiomics

CT based radiomic features are capable of separating patients with NSCLC [80][81][82][83][84] as well as head and neck squamous cell carcinoma (HNSCC) [81] with differing risk profiles for survival. Furthermore, CT based radiomic approaches allow prediction of dichotomous PD-L1 expression on tumor cells (tumor proportion score: TPS) [82][83] and density of CD3+ [82] or CD8+ [85] TILs in NSCLC. Successful anti-PD-1/anti-PD-L1 receptor-ligand-pair imaging by PET scans in mice with subcutaneously injected melanoma cells was demonstrated by Hettich et al. [86]. In a similar approach, Xing et al. [87] and Niemeijer et al. [88] investigated the correlation between PD-L1/PD-1 expression based on single photon emission computed tomography (SPECT), PD-L1/PD-1 PET and PD-L1/PD-1 expression assessed by immunohistochemistry (IHC) in NSCLC patients. Xing et al. used the anti-PD-L1 antibody NM-01, site-specifically labeled with technetium-99m, for SPECT imaging in 16 NSCLC patients (including squamous and non-squamous histology) in order to correlate tumor uptake with PD-L1 IHC. Patients with a PD-L1 expression ≤1% demonstrated statistically significantly lower tumor to peripheral blood tracer uptake ratios (mean 1.89 vs. 2.49, p = 0.0048) with a corresponding AUC of 0.88. It is noteworthy that four out of twelve patients with lymph nodes metastases showed considerable intra-patient differences (>20%) of PD-L1 expression [87]. Niemeijer et al. reported a statistically significant correlation between radiotracer uptake (18F-BMS-986192, standardized uptake value: SUV) and PD-L1 expression based on IHC (PD-L1 ≥50%: SUVpeak 8.2 versus PD-L1 <50%: SUVpeak 2.9, p = 0.018). The observed heterogeneous intrapatient and interpatient radiotracer uptake highlights the challenge to adequately assess tumor PD-L1 expression by core needle biopsies [88]. The latter two studies prove the feasibility to assess locoregional differences of PD-L1 expression in primary tumors and distant metastases. The assessment of intrapatient PD-L1 expression heterogeneity by radiomics may facilitate treatment decisions concerning intensity of therapy (ICI monotherapy versus ICI combined with chemotherapy) in clinical practice.

2.3. Radiomics Predict Clinical Outcome with ICI Therapy

By combining CT images and RNA-sequencing genomic data from tumor biopsies of patients with advanced solid tumors (MOSCATO trial) [89], Sun et al. developed a radiomic signature that could discriminate between high (>median) and low (<median) density of CD8+ TILs (AUC: 0.74,
p < 0.0001) [85] and validated the findings in three independent advanced solid tumor cohorts: TCGA validation set [90], tumor immune phenotype validation set [91] and immunotherapy-treated validation set [92]. Patients with a high radiomic score
(CD8+ TILs > median) showed a statistically significantly increased median OS (24.3 versus 11.5 months, p = 0.0081) in the immunotherapy-treated validation set and the radiomic score proved to be the strongest independent prognosticator for OS in multivariate analysis (hazard ratio (HR): 0.52,
p = 0.0022) [85]. Bensch et al. found a better correlation between clinical outcome and PD-L1 status assessment by PET imaging (89Zr-atezolizumab) in comparison to PD-L1 evaluation by IHC or RNA-sequencing data in 22 patients undergoing treatment with atezolizumab for bladder cancer, NSCLC or TNBC [93]. Khorrami et al. evaluated changes in the radiomic texture during two to three cycles of ICI therapy and reported the “delta-radiomic risk-score“ to predict response as well as OS with ICI in NSCLC [94]. Trebeschi et al. developed a radiomic signature based on pre-treatment CT images on a lesional level in advanced NSCLC and melanoma patients undergoing anti-PD-1 therapy. These radiomic features were significantly associated with response in pulmonary and nodal NSCLC metastases, whereas the model performed poorly on pulmonary and hepatic melanoma metastases. However, the model statistically significantly predicted OS in both tumor types (NSCLC: AUC: 0.76, p < 0.01; melanoma: AUC: 0.77, p < 0.01) [95]. Correlations of CT-based radiomic features and therapy response were also reported for patients with advanced ovarian cancer [96] and bladder cancer [97] undergoing immune-checkpoint blockade. Table 3 summarizes radiomics studies predicting clinical outcome with immune-checkpoint blockade.

Table 3. Prediction of clinical outcome by radiomics in cancer patients undergoing immune-checkpoint blockade.

Reference

Author

Tumor Entity

Findings

[85]

Sun et al.

solid tumors

OS prediction based on radiomics CD8+ cell score

[93]

Bensch et al.

bladder cancer, NSCLC, TNBC

↑ ORR/PFS/OS prediction by PET evaluation with zirconium-89-labeled atezolizumab compared to IHC or RNA-sequencing based PD-L1 assessment

[94]

Khorrami et al.

NSCLC

ORR and OS prediction based on changes in radiomic texture (“DelRADx”)

[95]

Trebeschi et al.

melanoma, NSCLC

Response prediction of individual metastases and OS prediction based on multiple radiomic features

[96]

Himoto et al.

ovarian cancer

Prediction of clinical benefit by intratumoral heterogeneity (radiomic feature) and by number of disease sites

[97]

Ligero et al.

solid tumors

↑ ORR prediction by clinical-radiomics signature score

[98]

Tunali et al.

NSCLC

Prediction of hyperprogressive disease based on clinical-radiomic models

[99]

Dercle et al.

non-squamous NSCLC

PFS prediction based on tumor volume reduction, infiltration of tumor boundaries or spatial heterogeneity

[100]

Korpics et al.

solid tumors

Prediction of local tumor failure, PFS and OS in cancer patients receiving SBRT and anti-PD-1 Tx based on a radiomics score

PET: positron emission tomography; PFS: progression-free survival; SBRT: stereotactic body radiotherapy, Tx: therapy; NSCLC: non-small cell lung cancer; TNBC: triple negative breast cancer; OS: overall survival; ORR: overall response rate; IHC: immunohistochemistry; PD-L1: programmed cell death-ligand 1;

On the one hand, a subset of advanced cancer patients derives long-term survival from immune-checkpoint blockade, on the other hand, up to nine per cent of patients experience hyperprogressive disease with rapid fatal outcome upon initiation of anti-PD-1/anti-PD-L1 therapy [101]. In a clinical-radiomic approach Tunali et al. were able to identify patients with a time to progression < 2 months or hyperprogressive disease within an advanced NSCLC cohort treated with single agent or double agent immunotherapy [[99]. The latter finding is of utmost importance in clinical practice as such cancer patients should not be treated with ICI monotherapy or with ICI at all. Apart from predicting clinical outcome with immunotherapy, radiomics also has the potential to predict immune-related adverse event. In a small series of 32 advanced cancer patients, Colen et al. found radiomic features that identified the two patients who experienced immunotherapy-induced pneumonitis (accuracy: 100%, p = 0.0033) [102].

The abovementioned findings corroborate the potential of radiomics to visualize drug distribution, tumor characteristics as well as tumor heterogeneity and the feasibility to predict clinical outcome with ICI. However, a major caveat remains the standardization of imaging acquisition, validation in prospective clinical trials and reproducibility in a real-world setting. ICI trials in advanced solid tumors such as the “INSPIRE“ trial (NCT02644369) are prospectively investigating changes in radiomic imaging parameters as well as correlations between tumor genomic profiles and radiomic imaging signatures.

References

  1. Daniel Chen; Ira Mellman; Elements of cancer immunity and the cancer–immune set point. Nature 2017, 541, 321-330, 10.1038/nature21349.
  2. Tao Jiang; Tao Shi; Henghui Zhang; Jie Hu; Yuanlin Song; Jia Wei; Shengxiang Ren; Caicun Zhou; Tumor neoantigens: from basic research to clinical applications.. Journal of Hematology & Oncology 2019, 12, 93, 10.1186/s13045-019-0787-5.
  3. Kaname Nosaki; Hideo Saka; Yukio Hosomi; Paul Baas; Gilberto De Castro; Martin Reck; Yi-Long Wu; Julie R. Brahmer; Enriqueta Felip; Takeshi Sawada; Kazuo Noguchi; Shi R. Han; Bilal Piperdi; Debra A. Kush; Gilberto Lopes; Safety and efficacy of pembrolizumab monotherapy in elderly patients with PD-L1–positive advanced non–small-cell lung cancer: Pooled analysis from the KEYNOTE-010, KEYNOTE-024, and KEYNOTE-042 studies. Lung Cancer 2019, 135, 188-195, 10.1016/j.lungcan.2019.07.004.
  4. Bin Wu; Fei Ma; Cost-effectiveness of adding atezolizumab to first-line chemotherapy in patients with advanced triple-negative breast cancer. Therapeutic Advances in Medical Oncology 2020, 12, , 10.1177/1758835920916000.
  5. Balar, A.V.; Castellano, D.; O’Donnell, P.H.; Grivas, P.; Vuky, J.; Powles, T.; Plimack, E.R.; Hahn, N.M.; de Wit, R.; Pang, L.; et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): A multicentre, single-arm, phase 2 study. Lancet Oncol. 2017, 18, 1483–1492.
  6. Burtness, B.; Harrington, K.J.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Baste, N.; Neupane, P.; Bratland, A.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  7. Jiaxin Zhu; Tiantian Zhang; Jiahao Li; Junming Lin; Wenhua Liang; Wenjie Huang; Ning Wan; Jie Jiang; Association Between Tumor Mutation Burden (TMB) and Outcomes of Cancer Patients Treated With PD-1/PD-L1 Inhibitions: A Meta-Analysis.. Frontiers in Pharmacology 2019, 10, 673, 10.3389/fphar.2019.00673.
  8. P C Doherty; R M Zinkernagel; A biological role for the major histocompatibility antigens.. The Lancet 1975, 1, 1406–1409.
  9. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al.et al. Faculty Opinions recommendation of Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors.. Science 2018, 359, 91–97, 10.3410/f.732063430.793542787.
  10. Florian Huemer; Gabriel Rinnerthaler; Theresa Westphal; Hubert Hackl; Georg Hutarew; Simon Peter Gampenrieder; Lukas Weiss; Richard Greil; Impact of antibiotic treatment on immune-checkpoint blockade efficacy in advanced non-squamous non-small cell lung cancer. Oncotarget 2018, 9, 16512-16520, 10.18632/oncotarget.24751.
  11. L. DeRosa; M.D. Hellmann; M. Spaziano; D. Halpenny; M. Fidelle; H. Rizvi; N. Long; A.J. Plodkowski; K.C. Arbour; J.E. Chaft; J.A. Rouche; L. Zitvogel; G. Zalcman; L. Albiges; B. Escudier; B. Routy; Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Annals of Oncology 2018, 29, 1437-1444, 10.1093/annonc/mdy103.
  12. Jonathan J. Havel; Diego Chowell; Timothy A. Chan; The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nature Reviews Cancer 2019, 19, 133-150, 10.1038/s41568-019-0116-x.
  13. Mark Yarchoan; Lee A. Albacker; Alexander C. Hopkins; Meagan Montesion; Karthikeyan Murugesan; Teena T. Vithayathil; Neeha Zaidi; Nilofer S. Azad; Daniel A. Laheru; Garrett M. Frampton; Elizabeth M. Jaffee; PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers.. JCI Insight 2019, 4, , 10.1172/jci.insight.126908.
  14. Samstein, R.M.; Lee, C.H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al.et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nature Genetics 2019, 51, 202-206, 10.1038/s41588-018-0312-8.
  15. C M Southam; Relationships of immunology to cancer: a review.. Cancer Research 1960, 20, , null.
  16. Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Sachet A. Shukla; Christian Blank; Lisa Zimmer; Antje Sucker; Uwe Hillen; Marnix H. Geukes Foppen; Simone M. Goldinger; et al.Jochen UtikalJessica C. HasselBenjamin WeideKatharina C. KaehlerCarmen LoquaiPeter MohrRalf GutzmerReinhard DummerStacey GabrielCatherine J. WuDirk SchadendorfLevi A. Garraway Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350, 207-211, 10.1126/science.aad0095.
  17. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al.et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463-1469, 10.1126/science.aaf1490.
  18. Maria R. Parkhurst; Paul F. Robbins; Eric Tran; Todd D. Prickett; Jared J. Gartner; Li Jia; Gabriel Ivey; Yong F. Li; Mona El-Gamil; Almin Lalani; et al.Jessica S. CrystalAbraham SachsEric M GrohSatyajit RayLien T. NgoScott KivitzAnna PasettoRami YossefFrank J. LoweryStephanie L. GoffWinifred LoGal CafriDrew C. DenigerParisa MalekzadehMojgan AhmadzadehJohn R. WunderlichRobert P.T. SomervilleSteven A. Rosenberg Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers. Cancer Discovery 2019, 9, 1022-1035, 10.1158/2159-8290.CD-18-1494.
  19. Michal Bassani-Sternberg; Sune Pletscher-Frankild; Lars Juhl Jensen; Matthias Mann; Mass spectrometry of human leukocyte antigen class I peptidomes reveals strong effects of protein abundance and turnover on antigen presentation.. Molecular & Cellular Proteomics 2015, 14, 658-73, 10.1074/mcp.M114.042812.
  20. Mahesh Yadav; Suchit Jhunjhunwala; Qui Phung; Patrick Lupardus; Joshua Tanguay; Stephanie Bumbaca; Christian Franci; Tommy K. Cheung; J. Fritsche; Toni Weinschenk; Zora Modrusan; Ira Mellman; Jennie R. Lill; Lelia Delamarre; Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 2014, 515, 572-576, 10.1038/nature14001.
  21. Anne-Mette Bjerregaard; Morten Nielsen; Vanessa Jurtz; Carolina Barra; S.R. Hadrup; Zoltan Szallasi; A.C. Eklund; An Analysis of Natural T Cell Responses to Predicted Tumor Neoepitopes. Frontiers in Immunology 2017, 8, 1566, 10.3389/fimmu.2017.01566.
  22. James Robinson; Jason A. Halliwell; James D. Hayhurst; Paul Flicek; Peter Parham; Steven G. E. Marsh; The IPD and IMGT/HLA database: allele variant databases.. Nucleic Acids Research 2014, 43, D423-431, 10.1093/nar/gku1161.
  23. .; The problem with neoantigen prediction. Nature Biotechnology 2017, 35, 97, 10.1038/nbt.3800.
  24. Hiroshi Mamitsuka; Predicting peptides that bind to MHC molecules using supervised learning of hidden markov models. Proteins: Structure, Function, and Bioinformatics 1998, 33, 460-474, 10.1002/(sici)1097-0134(19981201)33:4<460::aid-prot2>3.0.co;2-m.
  25. Donnes, P.; Elofsson, A. Prediction of MHC class I binding peptides, using SVMHC. BMC Bioinform. 2002, 3, 25
  26. Morten Nielsen; Claus Lundegaard; Peder Worning; Sanne Lise Lauemøller; Kasper Lamberth; Soren Buus; Søren Brunak; Ole Lund; Reliable prediction of T-cell epitopes using neural networks with novel sequence representations. Protein Science 2003, 12, 1007-1017, 10.1110/ps.0239403.
  27. Mette Voldby Larsen; Claus Lundegaard; Kasper Lamberth; Soren Buus; Søren Brunak; Ole Lund; Morten Nielsen; An integrative approach to CTL epitope prediction: A combined algorithm integrating MHC class I binding, TAP transport efficiency, and proteasomal cleavage predictions. European Journal of Immunology 2005, 35, 2295-2303, 10.1002/eji.200425811.
  28. Iris Antes; Shirley W. I. Siu; Thomas Lengauer; DynaPred: A structure and sequence based method for the prediction of MHC class I binding peptide sequences and conformations. Bioinformatics 2006, 22, e16-e24, 10.1093/bioinformatics/btl216.
  29. Nielsen, M.; Lundegaard, C.; Blicher, T.; Lamberth, K.; Harndahl, M.; Justesen, S.; Roder, G.; Peters, B.; Sette, A.; Lund, O.; et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS ONE 2007, 2, e796.
  30. Claus Lundegaard; Kasper Lamberth; Mikkel Harndahl; Soren Buus; Ole Lund; Morten Nielsen; NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8-11.. Nucleic Acids Research 2008, 36, W509-512, 10.1093/nar/gkn202.
  31. Ilka Hoof; Bjoern Peters; John Sidney; Lasse Eggers Pedersen; Alessandro Sette; Ole Lund; Soren Buus; Morten Nielsen; NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics 2008, 61, 1-13, 10.1007/s00251-008-0341-z.
  32. Hao Zhang; Ole Lund; Morten Nielsen; The PickPocket method for predicting binding specificities for receptors based on receptor pocket similarities: application to MHC-peptide binding. Bioinformatics 2009, 25, 1293-1299, 10.1093/bioinformatics/btp137.
  33. Yohan Kim; John Sidney; Clemencia Pinilla; Alessandro Sette; Bjoern Peters; Derivation of an amino acid similarity matrix for peptide:MHC binding and its application as a Bayesian prior. BMC Bioinformatics 2009, 10, 394-394, 10.1186/1471-2105-10-394.
  34. C. Lundegaard; Ole Lund; Morten Nielsen; Prediction of epitopes using neural network based methods. Journal of Immunological Methods 2010, 374, 26-34, 10.1016/j.jim.2010.10.011.
  35. Jorg J. A. Calis; Matt Maybeno; Jason A. Greenbaum; Daniela Weiskopf; Aruna D. De Silva; Alessandro Sette; Can Kesmir; Bjoern Peters; Properties of MHC Class I Presented Peptides That Enhance Immunogenicity. PLOS Computational Biology 2013, 9, e1003266, 10.1371/journal.pcbi.1003266.
  36. Pedersen, L.E.; Rasmussen, M.; Harndahl, M.; Nielsen, M.; Buus, S.; Jungersen, G. A combined prediction strategy increases identification of peptides bound with high affinity and stability to porcine MHC class I molecules SLA-1*04:01, SLA-2*04:01, and SLA-3*04:01. Immunogenetics 2016, 68, 157–165.
  37. Massimo Andreatta; Morten Nielsen; Gapped sequence alignment using artificial neural networks: application to the MHC class I system.. Bioinformatics 2015, 32, 511-7, 10.1093/bioinformatics/btv639.
  38. Morten Nielsen; Massimo Andreatta; NetMHCpan-3.0; improved prediction of binding to MHC class I molecules integrating information from multiple receptor and peptide length datasets. Genome Medicine 2016, 8, 33, 10.1186/s13073-016-0288-x.
  39. Shelly Kalaora; Eilon Barnea; Efrat Merhavi-Shoham; Nouar Qutob; Jamie K. Teer; Nilly Shimony; Jacob Schachter; Steven A. Rosenberg; Michal J. Besser; Arie Admon; et al.Yardena Samuels Use of HLA peptidomics and whole exome sequencing to identify human immunogenic neo-antigens. Oncotarget 2016, 7, 5110-5117, 10.18632/oncotarget.6960.
  40. Jurtz, V.; Paul, S.; Andreatta, M.; Marcatili, P.; Peters, B.; Nielsen, M. NetMHCpan-4.0: Improved Peptide-MHC Class I Interaction Predictions Integrating Eluted Ligand and Peptide Binding Affinity Data. J. Immunol. 2017, 199, 3360–3368.
  41. McGranahan, N.; Rosenthal, R.; Hiley, C.T.; Rowan, A.J.; Watkins, T.B.K.; Wilson, G.A.; Birkbak, N.J.; Veeriah, S.; Van Loo, P.; Herrero, J.; et al.et al. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 2017, 171, 1259-1271.e11, 10.1016/j.cell.2017.10.001.
  42. Marta Łuksza; Nadeem Riaz; Vladimir Makarov; Vinod P. Balachandran; Matthew D. Hellmann; Alexander Solovyov; Naiyer A. Rizvi; Taha Merghoub; Arnold J. Levine; Timothy A. Chan; et al.Jedd D. WolchokBenjamin Greenbaum A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517-520, 10.1038/nature24473.
  43. Timothy O'donnell; Alex Rubinsteyn; Maria Bonsack; Angelika B. Riemer; Uri Laserson; Jeffrey Hammerbacher; MHCflurry: Open-Source Class I MHC Binding Affinity Prediction. Cell Systems 2018, 7, 129-132.e4, 10.1016/j.cels.2018.05.014.
  44. S. Kim; Han Sang Kim; E. Kim; Min Goo Lee; E. C. Shin; Soonmyung Paik; Neopepsee: accurate genome-level prediction of neoantigens by harnessing sequence and amino acid immunogenicity information. Annals of Oncology 2018, 29, 1030-1036, 10.1093/annonc/mdy022.
  45. Agnes Ling; Anna Löfgren-Burström; Pär Larsson; Xingru Li; Maria L. Wikberg; Åke Öberg; Roger Stenling; Sofia Edin; Richard Palmqvist; TAP1 down-regulation elicits immune escape and poor prognosis in colorectal cancer. OncoImmunology 2017, 6, e1356143, 10.1080/2162402X.2017.1356143.
  46. Mark H. Einstein; Suzanne Leanza; Lydia G. Chiu; Nicolas F. Schlecht; Gary L. Goldberg; Bettie M. Steinberg; Robert D. Burk; Genetic variants in TAP are associated with high-grade cervical neoplasia.. Clinical Cancer Research 2009, 15, 1019-1023, 10.1158/1078-0432.CCR-08-1207.
  47. Gettinger, S.; Choi, J.; Hastings, K.; Truini, A.; Datar, I.; Sowell, R.; Wurtz, A.; Dong, W.; Cai, G.; Melnick, M.A.; et al.et al. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discovery 2017, 7, 1420-1435, 10.1158/2159-8290.CD-17-0593.
  48. Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al.et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nature Communications 2017, 8, 1136, 10.1038/s41467-017-01062-w.
  49. Barbara Schrörs; Silke Lübcke; Volker Lennerz; Martina Fatho; Anne Bicker; Catherine Wölfel; Patrick Derigs; Thomas Hankeln; Dirk Schadendorf; Annette Paschen; et al.Thomas Wölfel HLA class I loss in metachronous metastases prevents continuous T cell recognition of mutated neoantigens in a human melanoma model. Oncotarget 2017, 8, 28312-28327, 10.18632/oncotarget.16048.
  50. Schrors, B.; Lubcke, S.; Lennerz, V.; Fatho, M.; Bicker, A.; Wolfel, C.; Derigs, P.; Hankeln, T.; Schadendorf, D.; Paschen, A.; et al. HLA class I loss in metachronous metastases prevents continuous T cell recognition of mutated neoantigens in a human melanoma model. Oncotarget 2017, 8, 28312–28327.
  51. Lu Huang; Shruti Malu; Jodi A. McKenzie; Miles C. Andrews; Amjad H. Talukder; Trang Tieu; Tatiana V. Karpinets; Cara Haymaker; Marie-Andrée Forget; Leila J. Williams; et al.Zhe WangRina MbofungZhi-Qiang WangRichard Eric DavisRoger S. LoJennifer A. WargoMichael A. DaviesChantale BernatchezTimothy P. HeffernanRodabe N. AmariaAnil KorkutWeiyi PengJason RoszikGregory LizéeScott E. WoodmanPatrick Hwu The RNA-binding Protein MEX3B Mediates Resistance to Cancer Immunotherapy by Downregulating HLA-A Expression. Clinical Cancer Research 2018, 24, 3366-3376, 10.1158/1078-0432.ccr-17-2483.
  52. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al.et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. New England Journal of Medicine 2016, 375, 819-829, 10.1056/NEJMoa1604958.
  53. Sucker, A.; Zhao, F.; Pieper, N.; Heeke, C.; Maltaner, R.; Stadtler, N.; Real, B.; Bielefeld, N.; Howe, S.; Weide, B.; et al. Acquired IFNgamma resistance impairs anti-tumor immunity and gives rise to T-cell-resistant melanoma lesions. Nat. Commun. 2017, 8, 15440.
  54. Daniel Sanghoon Shin; Jesse M. Zaretsky; Helena Escuin-Ordinas; Angel Garcia-Diaz; Siwen Hu-Lieskovan; Anusha Kalbasi; Catherine S. Grasso; Willy Hugo; Salemiz Sandoval; Davis Y. Torrejon; Nicolaos Palaskas; Gabriel Abril Rodriguez; Giulia Parisi; Ariel Azhdam; Bartosz Chmielowski; Grace Cherry; Elizabeth Seja; Beata Berent-Maoz; I. Peter Shintaku; Dung Thi Le; Ew M. Pardoll; Luis A. Diaz; Paul C. Tumeh; Thomas G. Graeber; Roger S. Lo; Begoña Comin-Anduix; Antoni Ribas; Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations.. Cancer Discovery 2016, 7, 188-201, 10.1158/2159-8290.CD-16-1223.
  55. Lloyd Smith; Chowell D; Morris Lgt; Grigg Cm; Weber Jk; Samstein Rm; Makarov V; Kuo F; Kendall Sm; Requena D; et al.Riaz NGreenbaum BCarroll JGaron EHyman DmZehir ASolit DBerger MZhou RNaiyer A. RizviChan Ta Faculty Opinions recommendation of Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy.. Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature 2018, 359, 582–587, 10.3410/f.732240293.793542866.
  56. Aaron Goodman; Rachel Pyke; Shumei Kato; Ryosuke Okamura; Ethan Sokol; Garrett Michael Frampton; Vincent A. Miller; Hannah Carter; Razelle Kurzrock; MHC-1 genotype as a predictor of response to immunotherapy.. Journal of Clinical Oncology 2019, 37, 149, 10.1200/jco.2019.37.8_suppl.149.
  57. Alexander C. Hopkins; Mark Yarchoan; Jennifer N. Durham; Erik C. Yusko; Julie Rytlewski; Harlan S. Robins; Daniel A. Laheru; Dung T. Le; Eric R. Lutz; Elizabeth M. Jaffee; et al. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight 2018, 3, 122092, 10.1172/jci.insight.122092.
  58. Lídia Robert; Christina Harview; Ryan Emerson; Xiaoyan Wang; Stephen Mok; Blanca Homet; Begonya Comin-Anduix; Richard C Koya; Harlan Robins; Paul C Tumeh; et al.Antoni Ribas Distinct immunological mechanisms of CTLA-4 and PD-1 blockade revealed by analyzing TCR usage in blood lymphocytes. OncoImmunology 2014, 3, e29244, 10.4161/onci.29244.
  59. Sabrina A. Hogan; Anaïs Courtier; Phil F. Cheng; Nicoletta F. Jaberg-Bentele; Simone M. Goldinger; Manuarii Manuel; Solène Perez; Nadia Plantier; Jean-François Mouret; Thi Dan Linh Nguyen-Kim; et al.Marieke I.G. RaaijmakersPia KvistborgNicolas PasqualJohn B.A.G. HaanenReinhard DummerMitchell P. Levesque Peripheral Blood TCR Repertoire Profiling May Facilitate Patient Stratification for Immunotherapy against Melanoma. Cancer Immunology Research 2018, 7, 77-85, 10.1158/2326-6066.cir-18-0136.
  60. E Ghorani; R. Rosenthal; N. McGranahan; J.L. Reading; M. Lynch; K.S. Peggs; Charles Swanton; Sergio A. Quezada; Differential binding affinity of mutated peptides for MHC class I is a predictor of survival in advanced lung cancer and melanoma. Annals of Oncology 2018, 29, 271-279, 10.1093/annonc/mdx687.
  61. Lena Katharina Freudenmann; Ana Marcu; Stefan Stevanović; Mapping the tumour human leukocyte antigen (HLA) ligandome by mass spectrometry. Immunology 2018, 154, 331-345, 10.1111/imm.12936.
  62. Chowell, D.; Krishna, S.; Becker, P.D.; Cocita, C.; Shu, J.; Tan, X.; Greenberg, P.D.; Klavinskis, L.S.; Blattman, J.N.; Anderson, K.S. TCR contact residue hydrophobicity is a hallmark of immunogenic CD8+ T cell epitopes. Proc. Natl. Acad. Sci. USA 2015, 112, E1754–E1762.
  63. Alexandra Snyder; Vladimir Makarov; Taha Merghoub; Jianda Yuan; Jesse M. Zaretsky; Alexis Desrichard; Logan Walsh; Michael A. Postow; Phillip Wong; Teresa S. Ho; et al.Travis HollmannCameron BruggemanKasthuri KannanYanyun LiCeyhan ElipenahliCailian LiuChristopher T. HarbisonLisu WangAntoni RibasJedd D. WolchokTimothy A. Chan Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. New England Journal of Medicine 2014, 371, 2189-2199, 10.1056/NEJMoa1406498.
  64. Nick Van Dijk; Samuel A. Funt; Christian U. Blank; Thomas Powles; Jonathan E. Rosenberg; Michiel S Van Der Heijden; The Cancer Immunogram as a Framework for Personalized Immunotherapy in Urothelial Cancer. European Urology 2019, 75, 435-444, 10.1016/j.eururo.2018.09.022.
  65. Takahiro Karasaki; Kazuhiro Nagayama; Hideki Kuwano; Jun-Ichi Nitadori; Masaaki Sato; Masaki Anraku; Akihiro Hosoi; Hirokazu Matsushita; Yasuyuki Morishita; Kosuke Kashiwabara; Masaki Takazawa; Osamu Ohara; Kazuhiro Kakimi; Jun Nakajima; An Immunogram for the Cancer-Immunity Cycle: Towards Personalized Immunotherapy of Lung Cancer. Journal of Thoracic Oncology 2017, 12, 791-803, 10.1016/j.jtho.2017.01.005.
  66. Feng Xie; Jianjun Zhang; Jiayin Wang; Alexandre Reuben; Wei Xu; Xin Yi; Frederick S. Varn; Yongsheng Ye; Junwen Cheng; Miao Yu; et al.Yue WangYufeng LiuMingchao XiePeng DuKe MaXin MaPenghui ZhouShengli YangYaobing ChenGuoping WangXuefeng XiaZhongxing LiaoJohn HeymachIgnacio I. WistubaP. Andrew FutrealKai YeChao ChengTian Xia Multifactorial Deep Learning Reveals Pan-Cancer Genomic Tumor Clusters with Distinct Immunogenomic Landscape and Response to Immunotherapy. Clinical Cancer Research 2020, ., ., 10.1158/1078-0432.ccr-19-1744.
  67. Alexander Haragan; John Field; Michael P.A. Davies; Carles Escriu; Aaron M. Gruver; John R. Gosney; Heterogeneity of PD-L1 expression in non-small cell lung cancer: Implications for specimen sampling in predicting treatment response.. Lung Cancer 2019, 134, 79-84, 10.1016/j.lungcan.2019.06.005.
  68. Hui Yu; Theresa A. Boyle; Caicun Zhou; David L. Rimm; Fred R. Hirsch; PD-L1 Expression in Lung Cancer.. Journal of Thoracic Oncology 2016, 11, 964-975, 10.1016/j.jtho.2016.04.014.
  69. Carol C. Wu; Michael M. Maher; Jo-Anne O. Shepard; Complications of CT-Guided Percutaneous Needle Biopsy of the Chest: Prevention and Management. American Journal of Roentgenology 2011, 196, , 10.2214/ajr.10.4659.
  70. Pornpen Thampanitchawong; Teerha Piratvisuth; Liver biopsy:complications and risk factors.. World Journal of Gastroenterology 1999, 5, , null.
  71. Giuseppe L Banna; Timothée Olivier; Francesco Rundo; Umberto Malapelle; Filippo Fraggetta; Massimo Libra; Alfredo Addeo; The Promise of Digital Biopsy for the Prediction of Tumor Molecular Features and Clinical Outcomes Associated With Immunotherapy.. Frontiers in Medicine 2019, 6, 172, 10.3389/fmed.2019.00172.
  72. Yang, L.; Dong, D.; Fang, M.; Zhu, Y.; Zang, Y.; Liu, Z.; Zhang, H.; Ying, J.; Zhao, X.; Tian, J. Can CT-based radiomics signature predict KRAS/NRAS/BRAF mutations in colorectal cancer? Eur. Radiol. 2018, 28, 2058–2067.
  73. Huang, Q.Y.; Liu, Z. Radiomics approach to characterize microsatellite instability: A CT-based radiomic signature for the detection of the MSI-H phenotype in colorectal cancer. In Proceedings of the ECR 2019, Vienna, Austria, 27 February–3 March 2019.
  74. Stephen Sf Yip; John Kim; Thibaud P Coroller; Chintan Parmar; Emmanuel Rios Velazquez; Elizabeth Huynh; Raymond H Mak; Hugo J. W. L. Aerts; Associations Between Somatic Mutations and Metabolic Imaging Phenotypes in Non-Small Cell Lung Cancer.. Journal of Nuclear Medicine 2016, 58, 569-576, 10.2967/jnumed.116.181826.
  75. Olivier Gevaert; Sebastian Echegaray; Amanda Khuong; Chuong D. Hoang; Joseph B. Shrager; Kirstin C. Jensen; Gerald J. Berry; H. Henry Guo; Charles Lau; Sylvia K. Plevritis; Daniel L. Rubin; Sandy Napel; Ann N. Leung; Predictive radiogenomics modeling of EGFR mutation status in lung cancer. Scientific Reports 2017, 7, 41674, 10.1038/srep41674.
  76. R.S. Herbst; G. Lopes; D.M. Kowalski; K. Kasahara; Y.-L. Wu; G. De Castro; B.C. Cho; H.Z. Turna; R. Cristescu; D. Aurora-Garg; et al.J. LuncefordJ. KobieM. AyersM.C. PietanzaB. PiperdiT.S.K. Mok LBA4 Association of KRAS mutational status with response to pembrolizumab monotherapy given as first-line therapy for PD-L1-positive advanced non-squamous NSCLC in Keynote-042. Annals of Oncology 2019, 30, xi63-xi64, 10.1093/annonc/mdz453.001.
  77. Jordan Berlin; Le Dt; Durham Jn; Smith Kn; Wang H; Bartlett Br; Aulakh Lk; Lu S; Kemberling H; Wilt C; et al.Luber BsWong FAzad NsRucki AaLaheru DDonehower RZaheer AFisher GaCrocenzi TsLee JjGreten TfDuffy AgCiombor KkEyring AdLam BhJoe AKang SpHoldhoff MDanilova LCope LMeyer CZhou SGoldberg RmArmstrong DkBever KmFader AnTaube JHousseau FSpetzler DXiao NPardoll DmPapadopoulos NKinzler KwEshleman JrVogelstein BAnders RaDiaz La Faculty Opinions recommendation of Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade.. Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature 2020, 357, 409–413, 10.3410/f.727697784.793571316.
  78. Aaron Lisberg; Edward B. Garon; The Italian Nivolumab Expanded Access Program Confirms the Limitations of Single-Agent PD-1 Inhibition in EGFR-Mutant and Never-Smoking Patients with NSCLC. Journal of Thoracic Oncology 2018, 13, 1058-1059, 10.1016/j.jtho.2018.06.003.
  79. Chee Khoon Lee; Johnathan Man; Sarah J. Lord; Matthew Jon Links; Val Gebski; Tony S.K. Mok; James Chih-Hsin Yang; Checkpoint Inhibitors in Metastatic EGFR- Mutated Non–Small Cell Lung Cancer—A Meta-Analysis. Journal of Thoracic Oncology 2017, 12, 403-407, 10.1016/j.jtho.2016.10.007.
  80. Olya Grove; Anders E. Berglund; Matthew B. Schabath; Hugo J. W. L. Aerts; Aldo Dekker; Hua Wang; Emmanuel Rios Velazquez; Philippe Lambin; Yuhua Gu; Yoganand Balagurunathan; Edward Eikman; Robert A. Gatenby; Steven Eschrich; Robert J. Gillies; Quantitative Computed Tomographic Descriptors Associate Tumor Shape Complexity and Intratumor Heterogeneity with Prognosis in Lung Adenocarcinoma. PLOS ONE 2015, 10, e0118261, 10.1371/journal.pone.0118261.
  81. Hugo J. W. L. Aerts; Emmanuel Rios Velazquez; Ralph T. H. Leijenaar; Chintan Parmar; Patrick Grossmann; Sara Cavalho; Johan Bussink; René Monshouwer; Benjamin Haibe-Kains; Derek Rietveld; Frank Hoebers; Michelle M. Rietbergen; C. René Leemans; Aldo Dekker; John Quackenbush; Robert J. Gillies; Philippe Lambin; Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nature Communications 2014, 5, 4006, 10.1038/ncomms5006.
  82. Chad Tang; Brian Hobbs; Ahmed Amer; Xiao Li; Carmen Behrens; Jaime Rodriguez Canales; Edwin Parra Cuentas; Pamela Villalobos; David Fried; Joe Y. Chang; David S. Hong; James W. Welsh; Boris Sepesi; Laurence E. Court; Ignacio I. Wistuba; Eugene J. Koay; Development of an Immune-Pathology Informed Radiomics Model for Non-Small Cell Lung Cancer.. Scientific Reports 2018, 8, 1922, 10.1038/s41598-018-20471-5.
  83. Patil, P.D.; Bera, K.; Vaidya, P.; Prasanna, P.; Khunger, M.; Khunger, A.; Velcheti, V.; Madabhushi, A. Correlation of radiomic features with PD-L1 expression in early stage non-small cell lung cancer (ES-NSCLC) to predict recurrence and overall survival (OS). J. Clin. Oncol. 2018, 36.
  84. G. Mazzaschi; F. Quaini; G. Milanese; D. Madeddu; G. Bocchialini; L. Ampollini; L. Gnetti; C. Lagrasta; M. Silva; G. Roti; N. Sverzellati; M. Tiseo; Differentially regulated high-throughput CT imaging features correlate to distinct tumor immune contextures portraying a radiomic signature with prognostic impact on surgically resected NSCLC. Annals of Oncology 2019, 30, ii1, 10.1093/annonc/mdz072.001.
  85. Roger Sun; Elaine Johanna Limkin; Maria Vakalopoulou; Laurent Dercle; Stéphane Champiat; Shan Rong Han; Loic Verlingue; David Brandao; Andrea Lancia; Samy Ammari; et al.Antoine HollebecqueJean-Yves ScoazecAurélien MarabelleChristophe MassardJean-Charles SoriaCharlotte RobertNikos ParagiosÉric DeutschCharles Ferté A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study.. The Lancet Oncology 2018, 19, 1180-1191, 10.1016/S1470-2045(18)30413-3.
  86. Michael Hettich; Friederike Braun; Mark D. Bartholomä; Reinhold Schirmbeck; Gabriele Niedermann; High-Resolution PET Imaging with Therapeutic Antibody-based PD-1/PD-L1 Checkpoint Tracers. Theranostics 2016, 6, 1629-1640, 10.7150/thno.15253.
  87. Yan Xing; Gitasha Chand; Changchun Liu; Gary J.R. Cook; Jim O' Doherty; Lingzhou Zhao; Nicholas C. L. Wong; Levente K. Meszaros; Hong Hoi Ting; Jinhua Zhao; et al.Changcun Liu Early Phase I Study of a 99mTc-Labeled Anti-Programmed Death Ligand-1 (PD-L1) Single-Domain Antibody in SPECT/CT Assessment of PD-L1 Expression in Non-Small Cell Lung Cancer.. Journal of Nuclear Medicine 2019, 60, 1213-1220, 10.2967/jnumed.118.224170.
  88. A. N. Niemeijer; D. Leung; M. C. Huisman; I. Bahce; O. S. Hoekstra; G. A. M. S. Van Dongen; Ronald Boellaard; S. Du; W. Hayes; R. Smith; et al.Albert D. WindhorstN. H. HendrikseA. PootD. J. VugtsE. ThunnissenP. MorinD. LipovsekD. J. DonnellyS. J. BonacorsiL. M. VelasquezTanja D. De GruijlE. F. SmitA.J. De Langen Whole body PD-1 and PD-L1 positron emission tomography in patients with non-small-cell lung cancer. Nature Communications 2018, 9, 4664, 10.1038/s41467-018-07131-y.
  89. Christophe Massard; Stefan Michiels; Charles Ferté; Marie-Cécile Le Deley; Ludovic Lacroix; Antoine Hollebecque; Loic Verlingue; Ecaterina Ileana; Silvia Rosellini; Samy Ammari; et al.Maud Ngo-CamusRastislav BahledaAnas GazzahAndrea VargaSophie Postel-VinayYohann LoriotCaroline EvenIngrid BreuskinNathalie AugerBastien JobThierry De BaereFrederic DeschampsPhilippe VielhJean-Yves ScoazecVladimir LazarCatherine RichonVincent RibragÉric DeutschEric AngevinGilles VassalA.M.M. EggermontFabrice AndreJean-Charles Soria High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discovery 2017, 7, 586-595, 10.1158/2159-8290.cd-16-1396.
  90. John N. Weinstein; The Cancer Genome Atlas Research Network; Jun Li; Gordon B. Mills; Kenna R Mills Shaw; Brad A. Ozenberger; Kyle Ellrott; Ilya Shmulevich; Chris Sander; Joshua M. Stuart; et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nature Genetics 2013, 45, 1113-1120, 10.1038/ng.2764.
  91. Stéphane Champiat; Laurent Dercle; Samy Ammari; Christophe Massard; Antoine Hollebecque; Sophie Postel-Vinay; Nathalie Chaput; Alexander M Eggermont; Aurélien Marabelle; Jean-Charles Soria; et al.Charles Ferté Hyperprogressive disease (HPD) is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1. Clinical Cancer Research 2016, 23, 1920-1928, 10.1158/1078-0432.CCR-16-1741.
  92. Roger Sun; Stéphane Champiat; Laurent Dercle; Sandrine Aspeslagh; Eduardo Castanon; Elaine Johanna Limkin; Capucine Baldini; Sophie Postel-Vinay; Antoine Hollebecque; Christophe Massard; et al.Samy AmmariÉric DeutschJean-Charles SoriaAurélien MarabelleCharles Ferté Baseline lymphopenia should not be used as exclusion criteria in early clinical trials investigating immune checkpoint blockers (PD-1/PD-L1 inhibitors). European Journal of Cancer 2017, 84, 202-211, 10.1016/j.ejca.2017.07.033.
  93. Frederike Bensch; Elly L. Van Der Veen; Marjolijn N Lub-De Hooge; Annelies Jorritsma-Smit; Ronald Boellaard; Iris C. Kok; Sjoukje F. Oosting; Carolina P. Schröder; T. Jeroen N. Hiltermann; Anthonie J. Van Der Wekken; et al.H.J.M. GroenThomas C. KweeSjoerd G. EliasJourik GietemaSandra Sanabria BohorquezAlex De CrespignySimon-Peter WilliamsChristoph MancaoAdrienne H. BrouwersBernard FineElisabeth G.E. De Vries 89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nature Medicine 2018, 24, 1852-1858, 10.1038/s41591-018-0255-8.
  94. MohammadHadi Khorrami; Prateek Prasanna; Amit Gupta; Pradnya Patil; Priya D. Velu; Rajat Thawani; Germán Corredor; Mehdi Alilou; Kaustav Bera; Pingfu Fu; et al.Michael FeldmanVamsidhar VelchetiAnant Madabhushi Changes in CT Radiomic Features Associated with Lymphocyte Distribution Predict Overall Survival and Response to Immunotherapy in Non-Small Cell Lung Cancer.. Cancer Immunology Research 2019, 8, 108-119, 10.1158/2326-6066.CIR-19-0476.
  95. S. Trebeschi; S.G. Drago; Nicolai J. Birkbak; I. Kurilova; A.M. Cǎlin; Andrea Delli Pizzi; F. Lalezari; D.M.J. Lambregts; M.W. Rohaan; Chintan Parmar; et al.E.A. RozemanK.J. HarteminkC. SwantonJ B A G HaanenC.U. BlankE.F. SmitR.G.H. Beets-TanH J W L Aerts Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Annals of Oncology 2019, 30, 998-1004, 10.1093/annonc/mdz108.
  96. Yuki Himoto; Harini Veeraraghavan; Junting Zheng; Dmitriy Zamarin; Alexandra Snyder; Marinela Capanu; Stephanie Nougaret; Hebert A. Vargas; Fuki Shitano; Margaret Callahan; et al.Wei WangEvis SalaYulia Lakhman Computed Tomography–Derived Radiomic Metrics Can Identify Responders to Immunotherapy in Ovarian Cancer. JCO Precision Oncology 2019, ., 1-13, 10.1200/po.19.00038.
  97. M. Ligero; A. Garcia-Ruiz; C. Viaplana; M.V. Raciti; I. Matos; J Martín Liberal; C. Hierro; M. Gonzalez; R Morales Barrera; C. Suárez; et al.E. ElezI. BranaE. Muñoz-CouseloA. OakninE. FelipJ. TaberneroJ. CarlesR. DienstmannE. GarraldaR Perez Lopez Artificial intelligence combining radiomics and clinical data for predicting response to immunotherapy. Annals of Oncology 2019, 30, v476, 10.1093/annonc/mdz253.002.
  98. Ilke Tunali; Jhanelle E. Gray; Jin Qi; Mahmoud Abdalah; Daniel K. Jeong; Albert Guvenis; Robert J. Gillies; Matthew B. Schabath; Mahmoud Abdallah; Novel clinical and radiomic predictors of rapid disease progression phenotypes among lung cancer patients treated with immunotherapy: An early report.. Lung Cancer 2019, 129, 75-79, 10.1016/j.lungcan.2019.01.010.
  99. L. Dercle; M. Fronheiser; L. Lu; S. Du; W. Hayes; D.K. Leung; A. Roy; L.H. Schwartz; B. Zhao; Radiomic signatures for identification of tumors sensitive to nivolumab or docetaxel in squamous non-small cell lung cancer (sqNSCLC). Annals of Oncology 2019, 30, v773, 10.1093/annonc/mdz268.037.
  100. M.C. Korpics; S.R. Bhave; G. Redler; S. Pitroda; J.J. Luke; S.J. Chmura; A Validated Radiomics T Cell Score Predicts Response to Multi-site SBRT Combined with Pembrolizumab. International Journal of Radiation Oncology*Biology*Physics 2019, 104, 1189-1190, 10.1016/j.ijrobp.2019.05.044.
  101. Ignacio Matos; Juan Martin-Liberal; Alonso Garcia-Ruiz; Cinta Hierro; Maria Ochoa De Olza; Cristina Viaplana; Analía Azaro; María Vieito; Irene Brana; Gemma Mur; et al.Javier RosJose MateosGuillermo VillacampaRoger BerchéMafalda OliveiraMaria AlsinaElena ÉlezAna OakninEva Muñoz-CouseloJoan CarlesEnriqueta FelipJordi RodonJosep TaberneroRodrigo DienstmannRaquel Perez-LopezElena Garralda Capturing Hyperprogressive Disease with Immune-Checkpoint Inhibitors Using RECIST 1.1 Criteria. Clinical Cancer Research 2019, 26, 1846-1855, 10.1158/1078-0432.ccr-19-2226.
  102. Rivka R. Colen; Takeo Fujii; Mehmet A. Bilen; Aikaterini Kotrotsou; Srishti Abrol; K R Hess; Joud Hajjar; Maria E. Suarez-Almazor; Anas Alshawa; David S. Hong; et al.Dunia Giniebra-CamejoBettzy StephenVivek SubbiahAjay SheshadriTito MendozaSiqing FuPadmanee SharmaFunda Meric-BernstamAung Naing Radiomics to predict immunotherapy-induced pneumonitis: proof of concept. Investigational New Drugs 2017, 36, 601-607, 10.1007/s10637-017-0524-2.
More
Information
Subjects: Pathology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 771
Revisions: 3 times (View History)
Update Date: 29 Oct 2020
1000/1000