Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Purpose

Immunoprofiling to identify biomarkers and integration with clinical trial outcomes are critical to improving immunotherapy approaches for patients with cancer. However, the translational potential of individual studies is often limited by small sample size of trials and the complexity of immuno-oncology biomarkers. Variability in assay performance further limits comparison and interpretation of data across studies and laboratories.

Experimental design

To enable a systematic approach to biomarker identification and correlation with clinical outcome across trials, the Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC) Network was established through support of the Cancer MoonshotSM Initiative of the National Cancer Institute (NCI) and the Partnership for Accelerating Cancer Therapies (PACT) with industry partners via the Foundation for the NIH.

Results

The CIMAC-CIDC Network is composed of four academic centers with multidisciplinary expertise in cancer immunotherapy that perform validated and harmonized assays for immunoprofiling and conduct correlative analyses. A data coordinating center (CIDC) provides the computational expertise and informatics platforms for the storage, integration, and analysis of biomarker and clinical data.

Conclusions

This overview highlights strategies for assay harmonization to enable cross-trial and cross-site data analysis and describes key elements for establishing a network to enhance immuno-oncology biomarker development. These include an operational infrastructure, validation and harmonization of core immunoprofiling assays, platforms for data ingestion and integration, and access to specimens from clinical trials. Published in the same volume are reports of harmonization for core analyses: whole-exome sequencing, RNA sequencing, cytometry by time of flight, and IHC/immunofluorescence.

Free full text 


Logo of nihpaLink to Publisher's site
Clin Cancer Res. Author manuscript; available in PMC 2021 Oct 5.
Published in final edited form as:
PMCID: PMC8491462
NIHMSID: NIHMS1659034
PMID: 33419780

Network for biomarker immunoprofiling for cancer immunotherapy: Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC)

Abstract

Immunoprofiling to identify biomarkers and integration with clinical trials outcome are critical to improve immunotherapy approaches for cancer patients. However, the translational potential of individual studies is often limited by small sample size of trials and the complexity of immuno-oncology biomarkers. Variability in assays further limits comparison and interpretation of data across studies and laboratories.

To enable a systematic approach to biomarker identification and correlation with clinical outcome across trials, the Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC) Network was established through support of the Cancer MoonshotSM Initiative of the National Cancer Institute and the Partnership for Accelerating Cancer Therapies (PACT) with industry partners via the Foundation for the National Institutes of Health. The CIMAC-CIDC Network is composed of four academic centers (CIMACs) with multidisciplinary expertise in the field of cancer immunotherapy that provide validated and harmonized assays for immune profiling. A data coordinating center (CIDC) provides the computational expertise and resources for biomarker data storage and analysis platforms for correlation with clinical data.

This overview highlights strategies for assay harmonization to enable cross-trial and cross-site data analysis and describes key elements for establishing a network to enhance immuno-oncology biomarker development. These include an operational infrastructure; validation and harmonization of core immunoprofiling assays; platforms for data ingestion and integration; and access to specimens from clinical trials. Published in the same volume are reports of harmonization for core analyses: whole exome sequencing, RNA sequencing, cytometry by time of flight, and immunohistochemistry/immunofluorescence.

INTRODUCTION

Despite recent advances, the benefits of immunotherapy are limited to a minority of patients with cancer. While thousands of clinical trials (1) have been underway to test novel approaches and combination strategies, only a few investigational regimens have shown added benefit and received regulatory approvals. A major impediment to furthering the success of immunotherapy is inadequate understanding of the complex interplay between tumor and immune system, and the diverse mechanisms of resistance to therapy in individual patients. Studies of tumor and tumor microenvironment (TME) have the potential to improve understanding of tumor biology and drug action or resistance, and to identify biomarkers of response and toxicities. Especially, analysis of clinical samples at baseline, on treatment, and at progression should be able to enhance knowledge of the mechanism of immuno-oncology agents, inform new drug development and combination strategies, and provide biomarker strategies (2,3). The integration of stringently validated biomarkers in immunotherapy trials could accelerate therapeutic development and optimization of clinical outcome.

Findings in early- and late-phase clinical trials have led to identification of candidate predictive markers for response to anti-PD-1/PD-L1 monotherapy, such as PD-L1 expression (46), CD8+ T-cell density (7), tumor mutational burden (TMB) (8,9), neoantigen prediction (10), transcriptomic profiles (11), T-cell receptor (TCR) clonality (7), and microsatellite instability (MSI) status (12). Biomarkers associated with poor outcomes have also been identified; examples include tumor loss of antigen presentation machinery (13,14), activation of Wnt/beta-catenin signaling (15), and cyclin-dependent kinase 5 (CDK5) expression that may dampen ability of T cells to reject tumors (16). Furthermore, biomarkers to predict immune-related adverse events (irAEs) are also of high interest, particularly for irAEs with life-threatening consequences.

However, to date different biomarkers have been investigated with variable success and overall show no significant therapy association with response from inhibition of checkpoint pathways. Determining the predictive accuracy of biomarkers for immunotherapy must involve a comprehensive approach that encompasses the complexity of tumor biology and the host immune system. While multi-omics technologies are widely available to support objectives of biomarker discovery, variability in assay methodology, assay data reporting, and specimen collection and processing procedures prevents comparison and interpretation of data across individual laboratories and clinical trials (17).

Elements critical to a systematic effort in biomarker development across different sites and studies must include: multidisciplinary expertise and capacity for complex tumor and immune profiling; assay platforms that not only are analytically validated but also demonstrate comparable assay performance across laboratories; appropriate clinical study design and sufficient sample sizes to make statistically valid conclusions; and a database for biomarker and clinical data integration with bioinformatics tools for correlative analysis within and across trials (Figure 1).

An external file that holds a picture, illustration, etc.
Object name is nihms-1659034-f0001.jpg

Strategies to enhance the value of biomarker discovery through deep tumor-immune profiling on individual patients, biomarker analysis with clinical correlation within trials, and integrative analysis across trials.

METHODS

To facilitate the process of immune biomarker identification and comparison between different National Cancer Institute (NCI)-sponsored immunotherapy trials and laboratories, the NCI established the Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC) Network (https://cimac-network.org/; “the Network”). The Network is composed of four multidisciplinary academic centers (the CIMACs) with capacity for state-of-the-art immune profiling assays, and a data coordination center (CIDC) that provides a database and informatics platforms for analysis and integration of clinical and biomarker data across trials.

The CIMAC-CIDC Network was launched in September 2017 through the Cancer MoonshotSM initiative supported by the NCI. As the Network was being established, in parallel, it formed a collaboration with the Partnership for Accelerating Cancer Therapies (PACT), another Cancer MoonshotSM project, and it became the public side of the public-private partnership (PPP) overseen by the Foundation for the National Institutes of Health (FNIH). This collaboration of the Network and PACT, launched in February 2018 (18), allowed for exchange of ideas between the industry partners from twelve leading biopharmaceutical companies, the Food and Drug Administration (FDA), NCI, and the academic partners in the Network. The Network and PACT agreed that validation and harmonization of biomarkers are essential for the future of immunotherapy development. In particular, the industry partners, through the PPP, provide financial support for the bioinformatics needs of the Network for optimization of data collection methodologies, data integration, and building a database of biomarker and clinical data at the CIDC. Additionally, they support development of novel biomarker assays and correlative studies in immuno-oncology clinical trials sponsored by NCI, industry, and other organizations such as academic centers.

This overview describes components required for the establishment of the Network. Also published in this volume are separate manuscripts that summarize the harmonization efforts by the Network on key assay platforms, including those for whole exome sequencing (WES) and RNA sequencing (RNAseq); mass cytometry by time of flight (CyTOF); and singleplex and multiplex immunohistochemistry/immunofluorescence (IHC/IF).

THE CIMAC-CIDC NETWORK INFRASTRUCTURE

The four CIMACs are located at Dana-Farber Cancer Institute, the Icahn School of Medicine at Mount Sinai, the University of Texas MD Anderson Cancer Center, and Stanford University. The CIDC is hosted at Dana-Farber Cancer Institute. Each CIMAC encompasses a multidisciplinary group of investigators with basic, translational, clinical, and computational expertise required for conducting complex correlative analyses.

The operational structure of the Network is depicted in Figure 2. Clinical trial teams of the NCI- and PACT-solicited trials seeking to collaborate with the Network apply through an established process that includes evaluation of scientific merit and feasibility of biomarker studies in the context of the clinical trial. For the selected trials, the CIMAC-CIDC investigators partner with the clinical trial team to design a biomarker plan and conduct immunoprofiling assays and correlative analyses. For correlative studies sponsored by the private sector PACT funds, the industry members also form a working group that helps to advise and refine the study design with the trial team. Blood and tissue specimens from the trials are collected at or transferred to designated central biorepositories for pathology quality control, processing, and distribution to the CIMACs. Guidelines and template agreements were developed for data access and sharing, specimen transfers, and intellectual property requirements (https://cimac-network.org/documents/).

An external file that holds a picture, illustration, etc.
Object name is nihms-1659034-f0002.jpg

Organizational structure of the CIMAC-CIDC Network. Clinical trial teams supported by NCI or selected by PACT collaborate with the Network on the design and execution of correlative studies using specimens and data from clinical trials of immunotherapy. Assay data from CIMACs and clinical data from the trials are transferred to CIDC. CIMACs, trial investigators, and CIDC jointly perform integrative analyses of biomarker and clinical data using the CIDC bioinformatics platforms.

The four CIMACs provide a wide range of validated and harmonized assay platforms for comprehensive genomic, phenotypic, and functional characterizations for analysis of specimens from immunotherapy trials. Raw assay data generated by CIMAC laboratories are transferred to the CIDC. A set of clinical data elements is extracted from the clinical trial database and also transferred to the CIDC. CIDC functions as an immune-profiling data coordination center and facilitates Network activities through optimizing data collection methodologies, providing the central database investigator access, and ensuring bioinformatics tools for integrative data analysis of biomarker and clinical data, both within and across trials.

Currently over 30 clinical trials from various NCI trial networks, academic sites, and industry sponsors have been selected for collaboration with the CIMAC-CIDC Network. These trials range from phase I/pilot studies to randomized phase II and III trials and involve a variety of clinical settings, including pediatric malignancies, rare tumors, patients with preexisting autoimmune disorders, as well as patients with common solid tumors and hematological malignancies. Therapeutic strategies being tested in the trials include monotherapy with immunotherapy agents and combinations with other immune modulators, targeted agents, or chemotherapy/radiation. In addition to baseline tumor tissue and longitudinal blood sample collections, many early-phase trials also incorporate on-treatment and at-progression biopsies.

Overall, the Network has set out the following scientific and strategic goals:

  • Conduct both unbiased hypothesis-driven and hypothesis-generating correlative studies in immuno-oncology trials, with the goal of identifying candidate biomarkers and integrating with clinical trial outcome;

  • Establish the administrative infrastructure of the Network, including development of a CIMAC-CIDC Human Material Transfer Agreement (HMTA) involving multiple stakeholders (CIMAC-CIDC, NCI, clinical trial organizations and sponsors, and investigators), CIMAC-CIDC Guidelines to guide Network operations, and a study proposal intake process (documents found at https://cimac-network.org/documents/);

  • Develop and implement a specimen collection and processing “umbrella” protocol supporting the immune profiling assays of the CIMACs, including standardization of pre-analytical conditions “(found at https://cimac-network.org/documents/);

  • Analytically validate all Tier 1 and Tier 2 assays (Table 1) to be performed by CIMACs, conduct harmonization of the key assay platforms across different CIMACs, and establish reference standards for longitudinal monitoring of assay performance;

    Table 1.

    Tier 1, 2, and 3 assays in the CIMAC-CIDC Network

    Tier 1 assays (planned for all or most trials)Tier 2 assays (planned for selected trials)Tier 3 assays (highly novel and exploratory)
    • Whole Exome Sequencing (WES)
    • RNA sequencing (RNAseq)
    • The nCounter® platform for gene expression (Nanostring)
    • Multiplex immunohistochemistry/ immunofluorescence (mIHC/IF)
    • Singleplex IHC (sIHC)
    • Cytometry by Time Of Flight (CyTOF)
    • Olink immune-oncology immunoassay panel
    • Circulating tumor DNA (ctDNA)
    • Grand Serology ELISA
    • Multiplexed ion beam imaging (MIBI)
    • CyTOF Phosphoflow
    • Single-cell TCR-sequencing (scTCR-seq)
    • TCR-seq
    • Assay for Transposase-Accessible Chromatin (ATACseq)
    • Microbiome analysis (16S sequencing, shotgun metagenomics)
    • Single-cell RNA-sequencing (scRNAseq)
    • Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-Seq)
    • ELISPOT analysis for cytokine secreting cells
    • HLA tetramers for antigen-specific T cells
  • Support translational efforts in the immuno-oncology scientific community by providing access to a set of protocols for harmonized and validated assays that can be implemented by both academic and industrial laboratories;

  • Establish the CIDC, including the bioinformatics platforms for within- and cross-trial analysis and integration of biomarker and clinical data;

  • Facilitate broad data sharing with the larger research community by transferring data and findings from the CIDC to the NCI Cancer Research Data Commons (CRDC), including data from the industry sponsored trials;

  • Establish an evidence base that can be used by academia and industry for clinical validation of assays and biomarkers for trials.

SELECTION AND PRIORITIZATION OF BIOMARKER MODULES FOR CLINICAL TESTING

The CIMACs selected a variety of platforms to provide comprehensive tumor and immune profiling for characterization of anti-tumor immune responses. These assays are categorized by tiers based on scientific priority and technical robustness for implementation in clinical trials (Table 1).

Selection of assay platforms:

From a biological perspective, molecular profiling should encompass components essential to anti-tumor immune response, including tumor-intrinsic factors (e.g., immunogenicity, oncogenic pathways), host factors, and immune-cell subsets in the tumor microenvironment and periphery. A guiding principle for selection of CIMAC assays was to prioritize platforms that provide the most comprehensive and unbiased analysis.

CyTOF mass cytometry was selected for assessing function and phenotypes of immune-cell subsets, such as T cells, B cells, natural killer cells, macrophages, and myeloid-derived suppressor cells, as it was considered advantageous over flow cytometry for the higher number of biomarkers detected by antibody panels, with little or no spillover between detector channels (19). Olink was chosen as the core assay for profiling of cytokines, chemokines, and growth factors, which are essential to immune response and intercellular communication. Olink was selected after comparisons with several other platforms for multiplex soluble analyte measurement. The advantages of Olink include a dedicated panel for immuno-oncology, high number of measurable analytes per sample (over 90), high dynamic range of detection due to its proximity extension assay, low volume requirement, internal calibration controls, and good reproducibility. Whole exome sequencing (WES) was prioritized over use of targeted gene panel sequencing, to provide genomic correlates such as TMB calculated from single nucleotide variants (SNVs) (9), immunogenic neo-epitopes resulting from novel mutations (20), germline mutations and polymorphic variants (21), and MSI status (12). Whole transcriptome profiling (RNAseq) and TCR sequencing assays were chosen for their utility in measuring complex, dynamic physiological states and providing a wide range of information in a single readout, including tumor gene expression, neoantigen load, T-cell infiltrate, TCR clonality, HLA haplotype, and other signatures relevant to response or resistance to immunotherapy (22). The NanoString platform was identified as an alternative approach for transcriptional analysis. Although it covers a targeted panel of genes, it is robust, sensitive, and applicable especially in cases of low-quality RNA from FFPE samples. Tissue imaging via multiplex immunohistochemistry/Immunofluorescence (mIHC/mIF) was included as a core assay for its ability to probe multiple cellular markers simultaneously and provide information on spatial organization of cellular targets in relation to tumor cells, stroma, vasculature, and immune-cell subsets (23,24). Although not considered a “Tier 1” assay, microbiome analysis was harmonized between two CIMACs. Aspects harmonized included stool sample collection, aliquoting, DNA extraction, 16s rRNA sequencing, and computational considerations.

Prioritization of assays for implementation in clinical trials:

An organizing principle for biomarker prioritization was the concept of assay “tiers” based on level of comprehensive and unbiased features as well as the envisioned scope of assay application across clinical trials. To generate data that could be integrated across multiple trials, a core set of assays defined as “Tier 1” were chosen to be applied in all or most trials and require harmonization to ensure comparability of the data across CIMAC sites (Table 1).

“Tier 2” assays are used in selected trials and do not require harmonization; rather, they include assays less mature in their development and more appropriate for hypothesis generation. They could be available at a single CIMAC site and require less throughput than “Tier 1” assays. For example, the assay for transposase-accessible chromatin sequencing (ATACseq), which measures epigenetic changes in sorted specific immune-cell populations, is designated as a “Tier 2” assay (25). ATACseq analysis can be implemented in trials studying drugs that target DNA methylation or that work via other type of epigenetic reprogramming (26,27).

“Tier 3” assays are considered highly novel and under exploration, not necessarily ready for implementation in many trials. Examples include use of single-cell genome or single-cell transcriptome assays to provide a “deep dive” into immune system complexity. Such assays can reveal cell population differences, cellular evolutionary relationships, and clonal heterogeneity within the tumor (28).

Tier-based categorization can change over time as “Tier 2” assays prove sufficiently robust to become “Tier 1”. For example, the ctDNA assay, which yields information on tumor burden dynamics in cancer progression and may potentially circumvent the need for repeated tumor biopsies (29), has emerged as a biomarker of interest in multiple clinical trials and thus could be reclassified from “Tier 2” to “Tier 1”. With further development, “Tier 3” assays could potentially be promoted to “Tier 2” status.

CONSIDERATIONS FOR SPECIMEN COLLECTION AND PRE-ANALYTICS

How specimens are collected and processed for preservation can have a large impact on the quality of assay data and correlative analyses. The CIMAC-CIDC Network includes over 30 clinical studies led by multiple different trial groups, across a projected collection of 40,000 specimens from about 3,400 patients. Allocation of limited specimen material for various assays needed for comprehensive profiling across CIMAC sites frequently poses a logistical challenge.

Efforts to achieve the goals set for the CIMAC-CIDC Network require that specimens meet a high standard of quality to ensure robust and comparable profiling. To guide investigators through sample distribution options among a variety of assays and provide standardized methods for specimen collection and handling, the Network and NCI developed the Specimen Collection “Umbrella” protocol (found at https://cimac-network.org/documents/). The Umbrella protocol addresses various steps in the “sample flow” from tissue or blood sample acquisition at trial sites, to immediate processing and storage at biorepositories, to subsequent processing and downstream distribution to the CIMAC laboratories. An overview of the Umbrella is provided in Table 2.

Table 2.

Pre-analytical elements in the CIMAC specimen collection “Umbrella” protocol

Specimen TypeCollection and Processing at SiteImmediate Processing at BiobankProcessing at Biobank for Distribution to CIMAC LabsIntended Assay Use at CIMAC
Tissue biopsies
De Novo Core Needle Biopsy
Endoscopic/ Punch Biopsy
De Novo Surgical Resection
• 1–2 cores or 1 segment (FFPE)• Embed fixed tissue
• Store blocks
• Unstained slides + H&E
• DNA / RNA extraction
Fresh Frozen Samples:
 • WES/germline, RNA-Seq, TCR-Seq

FFPE Samples:
 • IF, IHC, MIBI, WES/germline, RNA-Seq, TCR-Seq
• 1–2 cores flash frozen or 1 segment flash frozen• Store frozen• DNA / RNA extraction
Archival FFPE Material• FFPE blocks or Unstained slides
• Core punches
• Store blocks or Vacuum-seal slides
• Refrigerate punches
• Unstained slides + H&E
• DNA / RNA extraction
Blood
Sodium Heparin Green-Top Tubes• 30 mL Draw• Isolate plasma and PBMCs
• Smart tubes
• Ship Smart tube, plasma, or PBMCs
• DNA (TCR-Seq)
• Plasma (Olink, ELISA)
• PBMCs (CyTOF, TCR-Seq)
Streck Cell-Free DNA Tubes• 10 mL Draw• Isolate plasma and Freeze aliquots• Ship plasma aliquots• cfDNA
K2-EDTA Purple-Top Tubes• 2 mL Draw (solid tumor germline)
• 5–10 mL Draw (hematologic germline)
• 2 mL Draw (TCR-Seq)
• Freeze germline aliquots
• 2 mL aliquots (TCR-Seq)
• Extract and ship DNA aliquots• Germline DNA, TCR-Seq
Bone marrow, CSF, stool
Bone Marrow Aspirates
OR
Cerebrospinal Fluid
• Custom volume in K2-EDTA tubes• Supernatant
• Cell fraction
• Ship aliquots
• Unstained slides + H&E
• DNA / RNA extraction
• CyTOF, Olink, IF, IHC, MIBI, RNA-Seq
Stool Samples• Self-collection (Ship ambient or frozen)• 2 mL aliquots (DNA-stabilizer)
• Frozen stool
• Ship frozen aliquots• 16S rRNA
• Shotgun metagenomics

FFPE: Formalin-fixed paraffin-embedded

The Umbrella protocol was developed using an iterative approach. Optimizing performance of CIMAC assays and defining specimen pre-analytic requirements were instrumental in the assay validation and harmonization efforts of the CIMAC-CIDC, described later in this overview. Existing biorepository standard operating procedures (SOPs) that had been well validated with clinical trial samples were employed or adapted as far as possible. Where feasible, novel approaches were considered to support the need for flexibility and maximize use of limited tissue.

Since its development, the Umbrella protocol has been incorporated into several trials selected for collaboration with CIMAC-CIDC. Potentially, the Umbrella could have broader applicability beyond CIMAC-CIDC, as a consensus guidance for prospective immunotherapy trials to ensure high-quality specimen collections for downstream analysis.

PRINCIPLES FOR ASSAY VALIDATION AND HARMONIZATION

To enable robust and systematic biomarker analysis across the CIMACs and across clinical trials, objective quality control measures are required for all assays to be performed in the Network. These measures include each assay’s analytical validity, including its pre-specified level of variability and reproducibility, concordance between laboratories, as well as acceptance criteria appropriate to its intended use (Table 3).

Table 3.

Analytical performance metrics evaluated for CIMAC assays

• Analytes
• Technical platform(s)
• Reagents, controls, and calibrators
• Quality control parameters for specimens/analytes (e.g., cell viability, RNA/protein quality/integrity)
• Critical pre-analytic variables
• Analytical performance characteristics for each assay:
   [black small square] Current status and results of studies defining the sensitivity, specificity, accuracy, precision, reproducibility, reportable range, reference ranges/intervals (normal values), turnaround time, and failure rate of the assay.
   [black small square] Use of positive and negative controls, calibrators, and reference standards.
   [black small square] Number of samples in the reproducibility study.
   [black small square] How run-to-run variation (Coefficient of Variation; CV) was assessed and handled.
   [black small square] How inter-laboratory variability in the measurements was assessed and how these sources of variation were minimized to maintain performance at all sites within acceptable limits and to prevent drift or bias in the assay.
   [black small square] Scoring procedures and type of data to be acquired:
     • Quantitative/continuously distributed
     • Semi-quantitative/ordered categorical
     • Qualitative/non-ordered categorical

Adapted from “Study Checklist for CTEP-Supported Early Phase Trials with Biomarker Assays”, found at http://ctep.cancer.gov/protocolDevelopment/ancillary_correlatives.htm

Analytical Validation:

An analytically validated assay should accurately and reliably measure the analyte of interest in specimens representative of the population of interest. Analytical validity is built on the concept of a total test, including pre-analytical, analytical, and interpretative/post-analytical phases of assay development (Table 3) (30). Analytical validation should demonstrate how robustly and reliably the test meets predefined performance standards of reproducibility, specificity, sensitivity, and dynamic range. Regression analysis by an appropriate linear or non-linear method should be performed comparing measured to expected biomarker assay performance across the quantification range. The general acceptance criteria for the correlation coefficient (r) should be predetermined based on the context of use (31).

The level of analytical validation required for CIMAC assays is set at the level of evidence claimed for research use only (RUO) assays, which are usually applied for sample characterization and hypothesis generation but cannot be used in clinical decision making and therefore are not required to be performed in a CLIA-certified laboratory. For the purpose of CIMAC-CIDC study, the assays went beyond the typical validation required for routine research-based assays, as the analytical validation process and consensus SOPs were aimed at informing future standardization and harmonization guidelines for these assays. Each “Tier 1” and “Tier 2” assay required a qualification document demonstrating analytical validation of the assay, including sensitivity, specificity, intra- and inter-assay precision, accuracy, linearity, reproducibility, and robustness/ruggedness (Table 3).

Assay harmonization for CIMAC-CIDC studies:

To allow comparisons of biomarker data across trials, concordance of these assays’ performance was established across the different CIMAC sites performing a given assay, following development of consensus protocols. To enable comparison and integration of assay data across different studies and sites, harmonization of laboratory-specific protocols and development of consensus SOPs are recommended. During this process, each participating laboratory evaluates and compares the validity of reagents, standards, methodologies, protocols, and data reporting specific to each laboratory. Development of consensus protocols enables data comparison and interpretation supporting biomarker development across different clinical trial sites (17) (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is nihms-1659034-f0003.jpg

Cross-site assay harmonization – An iterative process ensuring reproducibility and robustness of assays to overcome methodological and data variability across different sites. After Figure 1 in van der Burg SH et al. 2011 (17).

Across the CIMACs, the principles of harmonization have been applied and successfully completed for CIMAC Tier 1 assays assessing genomic (WES), transcriptomic (RNAseq), and phenotypic characterization of tumor (mIHC/mIF) and PBMC subtypes (CyTOF). Olink, also a Tier 1 assay, is validated and performed at a single CIMAC. The processes and results of the harmonization for individual assays are described in reports also published in this volume. All assays met the pre-defined acceptance criteria for concordance that had been established for each assay, demonstrating a high level of concordance of results between participating laboratories. The CIMAC assay SOPs are available on the CIMAC-CIDC website at https://cimac-network.org/assays/.

Statistical evaluation of assay performance:

Most biomarkers are measured as continuous variables, but some are categorical in nature. Repeatability is defined as the agreement of repeated measurements within one laboratory under similar conditions. On the other hand, harmonization is defined as the agreement of repeated measurements across different laboratories under various conditions. Because a large number of different biomarkers will be analyzed in a wide variety of settings, no single statistical method and no single criterion can be applied to analyze the agreement of measurements for all conditions. However, several statistical methods were applied to evaluate the agreement or concordance between measurements within and across laboratories:

  • Pearson and Spearman correlation coefficients are calculated to examine the agreement of two measurements of a continuous variable; a scatter plot can also be generated. The Pearson correlation coefficient is more efficient when the data are Gaussian-distributed. The Spearman correlation coefficient is more robust when the data deviate from the Gaussian distribution; it is less influenced by outliers. Although there is no uniformly accepted criterion of “acceptable” agreement, a correlation of greater than 0.7, 0.8, and 0.9 can be considered as having adequate, good, and excellent correlation, respectively.

  • Coefficient of variation (CV) is defined as the standard deviation of a measurement divided by its mean. By definition, it is a measure of variation on the scale of the mean. Hence, CV is a unitless measurement and is useful for quantifying the variation or precision of measurements. Both intra- and inter-laboratory CV’s can be calculated. Similarly, there is no uniformly accepted criterion regarding the magnitude of an “acceptable” CV. However, a CV of less than 0.3, 0.2, and 0.1 can be considered as having adequate, good, and excellent precision between the measurements, respectively.

  • Variance component model under the one-way analysis of variance can be constructed to model the variability both within and between laboratories in different sites. The total variability can be broken down into between-site variability, between-subject variability, and within-subject random error (i.e., measurement error). The relative magnitude of the different variabilities can be calculated by forming the percent of variability that is explained. Intraclass correlation coefficient (ICC) can also be calculated as the proportion of the total variance contributed by between-site variance. ICC can be generalized to allow for covariate effects. Small portions of variability due to site and due to random error indicate good harmonization.

  • Linear mixed effect model can be applied when biomarkers are measured over time within the same individual. Typically, the subject is considered as a random effect, and time a fixed effect. When multiple sites are involved, site can be added as a fixed or a random effect. When data are skewed, transformation can be applied to biomarkers to make the transformed values more Gaussian distributed. Other covariates can also be added as well. The contribution of various components to the biomarker value can be dissected and evaluated.

Reference materials for longitudinal assay performance monitoring:

To extend the full benefits of the CIMAC-CIDC validation and harmonization efforts, a long-term plan was put into place to monitor assay performance over the duration of the project. For several assays, standard reference materials were generated in batches for quality-control assessment of assay performance within and across different CIMACs over time (Table 4). Control materials will also be used in “bridging” studies to compare assay performance following a transition to a different platform or modification of an assay.

Table 4.

Longitudinal reference standards used by CIMAC-CIDC

Reference materialDescriptionAmount usedFrequency of testing
CyTOF BioLegend PBMCs 1:1 mixture of dual-labeled activated and resting PBMC Veri-Cells200–350 vials (1 million cells per vial)Spiked into every clinical sample at 10 % volume
Olink Various • Pooled plasma from healthy donors
• Randox cytokine cocktail
• Olink experimental controls
Over 1,000 aliquots availableUsed in every run
ELISA Grand Serology Healthy Plasma Pools • Healthy-donor plasma pools as negative control and titer calculations
• Positive plasma pools from patients with reactivity to several antigens
Over 300 mL of plasma available (3–15 µl per assay)Used as appropriate
IHC/IF MIBI CHTN Master TMA Master Tissue MicroArray (TMA) containing normal, neoplastic, and tumor tissueSequential sections will be distributed by CHTN (four TMAs blocks)Twice per year
WES Coriell Institute HapMap Cell Line Pool • Two pools of 10 HapMap cell lines containing different allele fractions.
• Two cell lines as germline controls
Cell-line pellets embedded into FFPE blocks, extracted DNA distributed to each siteTwice per year Used as analysis pipelines develop over time
Microbiome Various • Healthy donor fecal samples (RefA)
• ZymoBIOMICS Microbial Community Standard (RefB)
• DNA library of gut-relevant microbes of known abundance (RefC)
• RefA: 60 ready-to-use aliquots from ~100 mg of material
• RefB: commercially available
Used to control biases and batch effects for extractions, library prep, and sequencing runs

CHTN: Cooperative Human Tissue Network

THE CANCER IMMUNOLOGIC DATA COMMONS (CIDC)

The Dana-Farber Cancer Institute maintains and hosts the CIDC for the CIMAC-CIDC Network. The CIDC provides bioinformatics methods and the computational expertise and resources to facilitate the analysis of immuno-oncology trial data for the Network (Figure 4). CIDC will receive clinical data from various sources, including NCI trial network trials, investigator-initiated trials, and industry trials for integrative analysis of the assay and clinical data. While the integrity of the provided data is maintained, the data will be mapped to a clinical data model and current standards. In conjunction with the CIMACs, CIDC has developed data standards and software for recording molecular, clinical, and metadata generated by the Network. As these data standards evolve, the CIDC will work with the NCI Center for Cancer Data Harmonization (CCDH) to ensure data is available for rapid sharing via the NCI CRDC as well as facilitating future cross-trial analyses.

An external file that holds a picture, illustration, etc.
Object name is nihms-1659034-f0004.jpg

Network Data Coordination – CIDC provides the services and functionality below based on data received from CIMAC laboratories and Trial Organizations.

CIDC software:

The CIDC software platform is a Google cloud-based system designed to facilitate the ingestion of molecular, clinical, and metadata generated by CIMAC laboratories and participating clinical trial centers. The software system abides by stringent security controls under the Risk Management Framework published by the National Institute of Standards and Technology (NIST). The main components of the system are: a high-performance data transfer tool for CIMACs to transfer assay data and metadata to the cloud; centralized storage of all metadata in a managed database and files in cloud storage buckets; and a web-based data portal for browsing and downloading of data files associated with the clinical trials. Access management is implemented using a role-based methodology ensuring access is tightly controlled by the Network’s data access and sharing policies. The prototype CIDC data portal is operational and is in the process of ingesting assay data generated from clinical trial samples and different assay types.

CIDC bioinformatics:

The CIDC worked with the CIMACs to establish the experimental and computational pipelines and to identify the relevant pre- and post-analytical metadata to be collected with the assay data. Using a unified workflow management system, the CIDC has established several uniform bioinformatics processing pipelines. These include pipelines for processing WES, bulk RNA-seq, ATACseq, and TCR-seq data. The pipelines also provide self-contained, comprehensive HTML reports, and use conda and bioconda (32) to ensure reproducibility and portability.

The WES processing pipeline follows GATK (Gene Analysis Toolkit) best practices (33) implemented in Sentieon for identifying germline/somatic mutations, indels, and copy number variations. The pipeline also includes mutation interpretation, tumor purity, and clonality analysis features. Additionally, HLA typing and neoantigen prediction are incorporated specifically for immunological data.

The RNA-seq processing pipeline includes steps for preprocessing, quality control, conventional differential expression analysis, and downstream analysis (e.g., gene module and gene set enrichment analysis). Tailoring to immuno-oncology, it includes additional functions to estimate infiltrating immune cells, evaluate immunotherapy response prediction biomarkers, predict MSI status, infer infiltrating immune repertoires, and identify microbiota and their classifications. In addition, the CIDC has engaged in efforts to harmonize genomics data from three different experimental platforms (MD Anderson Cancer Center, Frederick National Laboratory for Cancer Research, and the Dana-Farber Cancer Institute) and is starting to use the pipelines to process trial samples. In the coming year, the CIDC aims to improve the neo-antigen prediction function by integrating WES and RNA-seq data and incorporating the newest immunopeptidome data.

For TCR-seq, the CIDC team built an interactive web application that generates HTML reports for users to visualize immune repertoire information for each sample, cluster the samples, and compare samples between different groups. The CIDC has also finished developing the ATACseq data processing pipeline based on a previous ChIP-seq pipeline (34). Finally, for CyTOF data, the CIDC licensed the Astrolabe platform, which uses an automated gating strategy to determine cell populations (35). All the bioinformatics pipelines developed and adopted are accompanied by documentation, software versions, analysis parameters, and reference data, and are tested regularly when necessary updates are made.

DISCUSSION

Assay harmonization was identified as an important objective for both the CIMAC-CIDC Network and the PACT PPP, to generate highly concordant and interpretable datasets across multiple laboratories and studies and to facilitate development of a database of biomarker and clinical data for secondary analyses. Harmonized assays reduce variability and enhance reproducibility of individual laboratory protocols and comparability of data across different laboratories and studies. In this regard, the primary objective of the Network activity has been achieved. In the second phase, the harmonized assays are being implemented for specimen analysis and correlation of assay data with clinical outcome variables. We hope the availability of CIMAC assay protocols and the publication of the data from CIMAC-CIDC Network harmonization projects will increase awareness in the immuno-oncology community of the importance of harmonization principles in successful biomarker identification, qualification, and implementation. It is the hope that these publicly available protocols will be adopted in academic and industry trials, allowing for uniform biomarker data generation enabling cross-trial analysis. Ultimately, the clinical utility of immune assays and optimization of immunotherapies based on biomarker data will depend on implementation of assay harmonization principles across the immuno-oncology community.

STATEMENT OF TRANSLATIONAL RELEVANCE

The CIMAC-CIDC is a Network of laboratories and a bioinformatics center established to perform biomarker analysis and correlation with clinical outcome data from immunotherapy trials. The specific goal for the Network is to perform comprehensive immune profiling of specimens from trials, using assays that span genomics, transcriptomics, and phenotyping analysis of the tumor, tumor microenvironment, and periphery. Identification of biomarkers to optimize immunotherapies for cancer patients requires analytically-validated and harmonized assays across multiple laboratories allowing cross-site and cross-trial analyses. Therefore, harmonization of assay protocols, a key requirement for reducing data variability and allowing interpretation and integration of assay data across trials and laboratories, plays an important part in the Network’s infrastructure. A centralized database for integration of clinical and assay data facilitates the identification of biomarkers to optimize immunotherapy approaches and management of cancer patients.

ACKNOWLEDGEMENTS

We would like to thank Jason Cristofaro, Anna Amar, Sherry Ansher, and Jianqiao Zhang at NCI and David Wholley and Lynn Smelkinson at FNIH for their dedicated work on the agreements for the Network. We would like to thank Irina Lubensky, Hala Makhlouf, and Chaz Stephens for their communications with the NCI biobanks; Melissa McKay-Daily, Nina Lukinova, and Tracy Lively for their roles in the incorporation of biomarker plans into clinical trial protocols and their overall guidance on the quality of biomarkers for clinical trials; Jeffrey Moscow and Percy Ivy for their guidance on matters related to the Experimental Therapeutics Clinical Trials Network (ETCTN); and Andrea Denicoff for overseeing the NCI contract support from the Emmes Company, LLC. We thank Jeremiah Faith and Jose Clemente for their work on microbiome harmonization. Importantly, we thank all of the CIMAC-CIDC and clinical collaborators for making the success of this important initiative possible. Special appreciation goes to the investigators at the NIH and the Frederick National Laboratory for Cancer Research, especially the Molecular Characterization Laboratory, who shared their expertise including serving as the reviewers of the validation and harmonization reports for the assays.

Scientific and financial support for the CIMAC-CIDC Network are provided through the National Cancer Institute (NCI) Cooperative Agreements: U24CA224319 (to S Gnjatic, D Del Valle, and G Bongers of the Icahn School of Medicine at Mount Sinai CIMAC), U24CA224331 (to FS Hodi, CJ Wu, and S Ranasinghe of the Dana-Farber Cancer Institute CIMAC), U24CA224285 (to JJ Lee, II Wistuba, G Al-ATrash, C Bernatchez, B Sanchez-Espiridion, RR Jenq, and C Chang of the MD Anderson Cancer Center CIMAC), U24CA224309 (to HT Maecker, S Bendall, and M Pichavant of the Stanford University CIMAC), and U24CA224316 (to XS Liu, E Cerami, J Lindsay, and J Yu of the CIDC at Dana-Farber Cancer Institute). MDACC also received support from NIH Cancer Center Support Grant P30CA016672 (to II Wistuba and JJ Lee) and the University of Texas SPORE NCI P50CA70907 (to II Wistuba). Additional support is made possible through the National Cancer Institute CTIMS Contract HHSN261201600002C (to RA Enos, M Bowman, VM Tatard-Leitman, and S Janssens of the Emmes Company, LLC).

Scientific and financial support for the Partnership for Accelerating Cancer Therapies (PACT) public-private partnership (PPP) are made possible through funding support provided to the FNIH by: AbbVie Inc., Amgen Inc., Boehringer-Ingelheim Pharma GmbH & Co. KG., Bristol-Myers Squibb, Celgene Corporation, Genentech Inc, Gilead, GlaxoSmithKline plc, Janssen Pharmaceutical Companies of Johnson & Johnson, Novartis Institutes for Biomedical Research, Pfizer Inc., and Sanofi.

Footnotes

CONFLICT OF INTEREST DISCLOSURE STATEMENT

Cathy Wu holds equity in BioNTech Inc and receives research funding from Pharmacyclics.

Shirley Liu is a cofounder, board and SAB member of GV20 Oncotherapy, SAB of 3DMed Care, consultant for Genentech, stock-holder of BMY, TMO, WBA, ABT, ABBV, and JNJ, and received research funding from Takeda and Sanofi.

Sacha Gnjatic reports consultancy and/or advisory roles for Merck, Neon Therapeutics, and OncoMed, and research funding from Bristol-Myers Squibb, Genentech, Immune Design, Agenus, Janssen R&D, Pfizer, Takeda, and Regeneron.

Ignacio I Wistuba is on the advisory Board of Genentech/Roche, Bayer, Bristol-Myers Squibb, Astra Zeneca/Medimmune, Pfizer, HTG Molecular, Asuragen, Merck, GlaxoSmithKline, Oncocyte, Guardant Health and MSD, is a speaker for Medscape, MSD, Genentech/Roche, Pfizer, AstraZeneca, Merck, and receives research support from Genentech, Oncoplex, HTG Molecular, DepArray, Merck, Bristol-Myers Squibb, Medimmune, Adaptive, Adaptimmune, EMD Serono, Pfizer, Takeda, Amgen, Karus, Johnson & Johnson, Bayer, Iovance, 4D, Novartis, and Akoya.

F Stephen Hodi has received grants, royalties, and clinical trial support from Bristol-Myers Squibb; is a consultant to Bristol-Myers Squibb, Merck, EMD Serono, Novartis, Genentech/Roche, Bayer, Aduro, Partners Therapeutics, Sanofi, Pfizer, Psioxus Therapeutics, 7 Hills Pharma, and Pieris Pharmaceutical; sits on advisory boards for Takeda, Compass Therapeutics, Apricity, Pionyr, Verastem, Torque, Rheos, Bicara, Celldex, Incyte, Corner Therapeutics, Hutchison MediPharma (US) Inc, Boehringer Ingelheim, and Amgen; holds equity in Apricity, Pionyr, Torque, Bicara; has patents pending for methods for treating MICA-related disorders (for which he has received royalties), angiopoiten-2 biomarkers predictive of anti-immune checkpoint response, PD-L1 isoforms for melanoma, methods of using pembrolizumab and trebananib, and anti-galectin antibody biomarkers predictive of anti-immune checkpoint and anti-angiogenesis responses; and holds patents on tumor antigens, therapeutic peptides, vaccine compositions and methods for restoring NKG2D pathway function against cancers, and antibodies to MHC class I polypeptiderelated sequence A.

Robert R Jenq has received consultant role fees from Merck, Karius, and Microbiome DX, advisory member role fees from Seres Therapeutics, Kaleido Biosciences, LISCure Biosciences, Maat Pharma, and Prolacta Bioscience, and patent licensing fees from Seres Biosciences.

Sean Bendall is a co-founder of IONPath and serves on its Board of Directors.

Helen X Chen, Minkyung Song, Holden T Maecker, David Patton, J Jack Lee, Stacey J Adam, Radim Moravec, Ethan Cerami, James Lindsay, Gheath Al-ATrash, Chantale Bernatchez, Stephen Hewitt, Elad Sharon, Howard Streicher, Rebecca A Enos, Melissa Bowman, Valerie M Tatard-Leitman, Beatriz Sanchez-Espiridion, Srinika Ranasinghe, Mina Pichavant, Diane Del Valle, Joyce Yu, Sylvie Janssens, Jenny Peterson-Klaus, Cathy Rowe, Gerold Bongers, Chia-Chi Chang, Jeffrey Abrams, Margaret M Mooney, James H Doroshow, Lyndsay N Harris, and Magdalena Thurin declare no conflict of interest.

REFERENCES

1. Xin Yu J, Hubbard-Lucey VM, Tang J. Immuno-oncology drug development goes global. Nat Rev Drug Discov 2019;18(12):899–900 doi 10.1038/d41573-019-00167-9. [Abstract] [CrossRef] [Google Scholar]
2. Roh W, Chen PL, Reuben A, Spencer CN, Prieto PA, Miller JP, et al.Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci Transl Med 2017;9(379) doi 10.1126/scitranslmed.aah3560. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
3. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017;168(4):707–23 doi 10.1016/j.cell.2017.01.017. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, et al.Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J Clin Oncol 2016;34(34):4102–9 doi 10.1200/JCO.2016.67.2477. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al.Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372(21):2018–28 doi 10.1056/NEJMoa1501824. [Abstract] [CrossRef] [Google Scholar]
6. Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al.PD-L1 Immunohistochemistry Assays for Lung Cancer: Results from Phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project. J Thorac Oncol 2017;12(2):208–22 doi 10.1016/j.jtho.2016.11.2228. [Abstract] [CrossRef] [Google Scholar]
7. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al.PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515(7528):568–71 doi 10.1038/nature13954. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
8. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al.Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9(1):34 doi 10.1186/s13073-017-0424-2. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
9. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al.Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371(23):2189–99 doi 10.1056/NEJMoa1406498. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
10. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al.Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351(6280):1463–9 doi 10.1126/science.aaf1490. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
11. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al.IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127(8):2930–40 doi 10.1172/JCI91190. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
12. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al.Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357(6349):409–13 doi 10.1126/science.aan6733. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Yoshihama S, Roszik J, Downs I, Meissner TB, Vijayan S, Chapuy B, et al.NLRC5/MHC class I transactivator is a target for immune evasion in cancer. Proc Natl Acad Sci U S A 2016;113(21):5999–6004 doi 10.1073/pnas.1602069113. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al.Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med 2016;375(9):819–29 doi 10.1056/NEJMoa1604958. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015;523(7559):231–5 doi 10.1038/nature14404. [Abstract] [CrossRef] [Google Scholar]
16. Dorand RD, Nthale J, Myers JT, Barkauskas DS, Avril S, Chirieleison SM, et al.Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity. Science 2016;353(6297):399–403 doi 10.1126/science.aae0477. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. van der Burg SH, Kalos M, Gouttefangeas C, Janetzki S, Ottensmeier C, Welters MJ, et al.Harmonization of immune biomarker assays for clinical studies. Sci Transl Med 2011;3(108):108ps44 doi 10.1126/scitranslmed.3002785. [Abstract] [CrossRef] [Google Scholar]
18. Baker RG, Hoos AX, Adam SJ, Wholley D, Doroshow JH, Lowy DR, et al.The Partnership for Accelerating Cancer Therapies. Cancer J 2018;24(3):111–4 doi 10.1097/PPO.0000000000000321. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Olsen LR, Leipold MD, Pedersen CB, Maecker HT. The anatomy of single cell mass cytometry data. Cytometry A 2019;95(2):156–72 doi 10.1002/cyto.a.23621. [Abstract] [CrossRef] [Google Scholar]
20. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al.Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348(6230):124–8 doi 10.1126/science.aaa1348. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Queirolo P, Morabito A, Laurent S, Lastraioli S, Piccioli P, Ascierto PA, et al.Association of CTLA-4 polymorphisms with improved overall survival in melanoma patients treated with CTLA-4 blockade: a pilot study. Cancer Invest 2013;31(5):336–45 doi 10.3109/07357907.2013.793699. [Abstract] [CrossRef] [Google Scholar]
22. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al.Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016;165(1):35–44 doi 10.1016/j.cell.2016.02.065. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
23. Parra ER, Uraoka N, Jiang M, Cook P, Gibbons D, Forget MA, et al.Validation of multiplex immunofluorescence panels using multispectral microscopy for immune-profiling of formalin-fixed and paraffin-embedded human tumor tissues. Sci Rep 2017;7(1):13380 doi 10.1038/s41598-017-13942-8. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Remark R, Merghoub T, Grabe N, Litjens G, Damotte D, Wolchok JD, et al.In-depth tissue profiling using multiplexed immunohistochemical consecutive staining on single slide. Sci Immunol 2016;1(1):aaf6925 doi 10.1126/sciimmunol.aaf6925. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
25. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods 2013;10(12):1213–8 doi 10.1038/nmeth.2688. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Marwitz S, Scheufele S, Perner S, Reck M, Ammerpohl O, Goldmann T. Epigenetic modifications of the immune-checkpoint genes CTLA4 and PDCD1 in non-small cell lung cancer results in increased expression. Clin Epigenetics 2017;9:51 doi 10.1186/s13148-017-0354-2. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Seremet T, Koch A, Jansen Y, Schreuer M, Wilgenhof S, Del Marmol V, et al.Molecular and epigenetic features of melanomas and tumor immune microenvironment linked to durable remission to ipilimumab-based immunotherapy in metastatic patients. J Transl Med 2016;14(1):232 doi 10.1186/s12967-016-0990-x. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
28. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, et al.Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 2019;176(1–2):404 doi 10.1016/j.cell.2018.12.034. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Goldberg SB, Narayan A, Kole AJ, Decker RH, Teysir J, Carriero NJ, et al.Early Assessment of Lung Cancer Immunotherapy Response via Circulating Tumor DNA. Clin Cancer Res 2018;24(8):1872–80 doi 10.1158/1078-0432.CCR-17-1341. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, et al.Validation of biomarkers to predict response to immunotherapy in cancer: Volume I - pre-analytical and analytical validation. J Immunother Cancer 2016;4:76 doi 10.1186/s40425-016-0178-1. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. Piccoli SP AB, Allinson J, Arnold M, Amur S, Aubrecht J, et al.Points to Consider Document: Scientific and Regulatory Considerations for the Analytical Validation of Assays Used in the Qualification of Biomarkers in Biological Matrices. Biomarker Assay Collaborative Evidentiary Considerations Writing Group, Critical Path Institute (C-Path) 2019. [Google Scholar]
32. Gruning B, Dale R, Sjodin A, Chapman BA, Rowe J, Tomkins-Tinch CH, et al.Bioconda: sustainable and comprehensive software distribution for the life sciences. Nat Methods 2018;15(7):475–6 doi 10.1038/s41592-018-0046-7. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
33. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al.From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics 2013;43:11 0 1– 0 33 doi 10.1002/0471250953.bi1110s43. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
34. Qin Q, Mei S, Wu Q, Sun H, Li L, Taing L, et al.ChiLin: a comprehensive ChIP-seq and DNase-seq quality control and analysis pipeline. BMC Bioinformatics 2016;17(1):404 doi 10.1186/s12859-016-1274-4. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Amir ED, Lee B, Badoual P, Gordon M, Guo XV, Merad M, et al.Development of a Comprehensive Antibody Staining Database Using a Standardized Analytics Pipeline. Front Immunol 2019;10:1315 doi 10.3389/fimmu.2019.01315. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/97369198
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/97369198

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1158/1078-0432.ccr-20-3241

Supporting
Mentioning
Contrasting
0
10
0

Article citations


Go to all (15) article citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

CIMAC (3)

CTIMS (1)

Dana-Farber Cancer Institute

    G Bongers of the Icahn School of Medicine at Mount Sinai CIMAC (1)

    LLC

      MD Anderson Cancer Center

        NCI NIH HHS (8)

        Stanford University