Report on computational assessment of Tumor Infiltrating Lymphocytes from the International Immuno-Oncology Biomarker Working Group (original) (raw)

Introduction

Very large adjuvant trials have illustrated how the current schemes fail to stratify patients with sufficient granularity to permit optimal selection for clinical trials, likely owing to application of an overly limited set of clinico-pathologic features1,2. Histologic evaluation of tumor-infiltrating lymphocytes (TILs) is emerging as a promising biomarker in solid tumors and has reached level IB-evidence as a prognostic marker in triple-negative (TNBC) and HER2-positive breast cancer3,4,5. Recently, the St Gallen Breast Cancer Expert Committee endorsed routine assessment of TILs for TNBC patients6. In the absence of adequate standardization and training, visual TILs assessment (VTA) is subject to a marked degree of ambiguity and interobserver variability7,8,9. A series of published guidelines from this working group (also known as TIL Working group or TIL-WG) aimed to standardize VTA in solid tumors, to improve reproducibility and clinical adoption10,11,12. TIL-WG is an international coalition of pathologists, oncologists, statisticians, and data scientists that standardize the assessment of Immuno-Oncology Biomarkers to aid pathologists, clinicians, and researchers in their research and daily practice. The value of these guidelines was highlighted in two studies systematically examining VTA reproducibility7,13. Nevertheless, VTA continues to have inherent limitations that cannot be fully addressed through standardization and training, including: 1. visual assessment will always have some degree of inter-reader variability; 2. the time constraints of routine practice make comprehensive assessment of large tissue sections challenging7,13; 3. perceptual limitations may introduce bias in VTA, for example, the same TILs density is perceived to be higher if there is limited stroma.

Research in using machine learning (ML) algorithms to analyze histology has recently produced encouraging results, fueled by improvements in both hardware and methodology. Algorithms that learn patterns from labeled data, based on “deep learning” neural networks, have obtained promising results in many challenging problems. Their success has translated well to digital pathology, where they have demonstrated outstanding performance in tasks like mitosis detection, identification of metastases in lymph node sections, tissue segmentation, prognostication, and computational TILs assessment (CTA)14,[15](#ref-CR15 "Tellez, D. et al. Whole-slide mitosis detection in H&E breast histology using PHH3 as a reference to train distilled stain-invariant convolutional networks. IEEE Trans. Med. Imaging (2018). https://doi.org/10.1109/TMI.2018.2820199

            "),[16](#ref-CR16 "Amgad, M. et al. Structured crowdsourcing enables convolutional segmentation of histology images. Bioinformatics 35, 3461–3467 (2019)."),[17](/articles/s41523-020-0154-2#ref-CR17 "Mobadersany, P. et al. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc. Natl Acad. Sci. USA 115, E2970–E2979 (2018)."). ‘Traditional' computational analysis of histology focuses on complex image analysis routines, that typically require extraction of handcrafted features and that often do not generalize well across data sets[18](/articles/s41523-020-0154-2#ref-CR18 "Litjens, G. et al. A survey on deep learning in medical image analysis. Med. Image Anal. 42, 60–88 (2017)."),[19](/articles/s41523-020-0154-2#ref-CR19 "Janowczyk, A. & Madabhushi, A. Deep learning for digital pathology image analysis: a comprehensive tutorial with selected use cases. J. Pathol. Inform. 7, 29 (2016)."). Although studies utilizing deep learning-based methods suggest impressive diagnostic performance, and better generalization across data sets, these methods remain experimental. Table [1](/articles/s41523-020-0154-2#Tab1) shows a sample of published CTA algorithms and discusses their strengths and limitations, in complementarity with a previous literature review by the TIL-WG[16](/articles/s41523-020-0154-2#ref-CR16 "Amgad, M. et al. Structured crowdsourcing enables convolutional segmentation of histology images. Bioinformatics 35, 3461–3467 (2019)."),[20](#ref-CR20 "Klauschen, F. et al. Scoring of tumor-infiltrating lymphocytes: from visual estimation to machine learning. Semin. Cancer Biol. 52, 151–157 (2018)."),[21](#ref-CR21 "Barua, S. et al. Spatial interaction of tumor cells and regulatory T cells correlates with survival in non-small cell lung cancer. Lung Cancer 117, 73–79 (2018)."),[22](#ref-CR22 "Schalper, K. A. et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J. Natl. Cancer Inst. 107, dju435 (2015)."),[23](#ref-CR23 "Brown, J. R. et al. Multiplexed quantitative analysis of CD3, CD8, and CD20 predicts response to neoadjuvant chemotherapy in breast cancer. Clin. Cancer Res. 20, 5995–6005 (2014)."),[24](#ref-CR24 "Saltz, J. et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep. 23, 181–193.e7 (2018)."),[25](#ref-CR25 "Amgad, M. et al. Joint region and nucleus segmentation for characterization of tumor infiltrating lymphocytes in breast cancer. in Medical Imaging 2019: Digital Pathology (eds. Tomaszewski, J. E. & Ward, A. D.) 20 (SPIE, 2019)."),[26](#ref-CR26 "Yuan, Y. et al. Quantitative image analysis of cellular heterogeneity in breast tumors complements genomic profiling. Sci. Transl. Med. 4, 157ra143 (2012)."),[27](#ref-CR27 "Basavanhally, A. N. et al. Computerized image-based detection and grading of lymphocytic infiltration in HER2+ breast cancer histopathology. IEEE Trans. Biomed. Eng. 57, 642–653 (2010)."),[28](#ref-CR28 "Corredor, G. et al. Spatial architecture and arrangement of tumor-infiltrating lymphocytes for predicting likelihood of recurrence in early-stage non-small cell lung cancer. Clin. Cancer Res. 25, 1526–1534 (2019)."),[29](#ref-CR29 "Yoon, H. H. et al. Intertumoral heterogeneity of CD3 and CD8 T-cell densities in the microenvironment of dna mismatch-repair-deficient colon cancers: implications for prognosis. Clin. Cancer Res. 25, 125–133 (2019)."),[30](#ref-CR30 "Swiderska-Chadaj, Z. et al. Convolutional Neural Networks for Lymphocyte detection in Immunohistochemically Stained Whole-Slide Images. (2018)."),[31](/articles/s41523-020-0154-2#ref-CR31 "Heindl, A. et al. Relevance of spatial heterogeneity of immune infiltration for predicting risk of recurrence after endocrine therapy of ER+ breast cancer. J. Natl. Cancer Inst. 110, djx137 (2018).").

Table 1 Sample CTA algorithms from the published literature.

Full size table

This review and perspective provides a broad outline of key issues that impact the development and translation of computational tools for TILs assessment. The ideal intended outcome is that CTA is successfully integrated into the routine clinical workflow; there is significant potential for CTA to address inherent limitations in VTA, and partially to mitigate high clinical demands in remote and under-resourced settings. This is not too difficult to conceive, and there are documented success stories in the commercialization and clinical adoption of computational algorithms including pap smear cytology analyzers32, blood analyzers33, and automated immunohistochemistry (IHC) workflows for ER, PR, Her2, and Ki6734,35,36,37,38.

The impact of staining approach on algorithm design and deployment

The type of stain and imaging modality will have a significant impact on algorithm design, validation, and capabilities. VTA guideline from the TIL-WG focus on assessment of stromal TILs (sTIL) using hematoxylin and eosin (H&E)-stained formalin-fixed paraffin-embedded sections, given their practicality and widespread availability, and the clear presentation of tissue architecture this stain provides10,11,12,39. Multiple studies have relied on in situ approaches like IHC, in situ hybridization (ISH), or genomic deconvolution in assessing TILs11,40,41. These modalities, however, are not typically used in daily clinical TILs assessment, as they are either still experimental, rely on assays of variable reliability, or involve stains not widely used in clinical practice, especially in low-income settings4,10,11. It is also difficult to quantitate and establish consistent thresholds for IHC measurement of even well-defined epitopes, such as Ki67 and ER, between different labs42,43. Moreover, there is no single IHC stain that highlights all mononuclear cells with high sensitivity and specificity, so H&E remains the stain typically used in the routine clinical setting44.

Despite these issues, there are significant potential advantages for using IHC with CTAs. By specifically staining TILs, IHC can make image analysis more reliable, and can also present new opportunities for granular TILs subclassification; different TIL subpopulations, including CD4+ T cells, CD8+ T cells, Tregs, NK cells, B cells, etc, convey pertinent information on immune activation and repression4,12. IHC is already utilized in standardization efforts for TILs assessment in colorectal carcinomas45,46. The specific highlighting of TILs by IHC can improve algorithm specificity47,48, and enable characterization of TIL subpopulations that have potentially distinct prognostic or predictive roles49,50. IHC can reduce misclassification of intratumoral TILs, which are difficult to reliably assess given their resemblance to tumor or supporting cells in many contexts like lobular breast carcinomas, small-blue-cell tumors like small cell lung cancer, and primary brain tumors4,12.

Characteristics of CTA algorithms that capture clinical guidelines

TIL-WG guidelines for VTA are somewhat complex4,10,11,12. There are VTA guidelines for many primary solid tumors and metastatic tumor deposits10,12, for untreated infiltrating breast carcinomas11, post-neoadjuvant residual carcinomas of the breast39, and for carcinoma in situ of the breast39. TILs score is defined as the fraction of a tissue compartment that is occupied by TILs (lymphoplasmacytic infiltrates). Different compartments have different prognostic relevance; tumor-associated sTILs is the most relevant in most solid tumors, whereas intratumoral TIL score (iTILs) has been reported to be prognostic, most notably in melanoma10. The spatial and visual context of TILs is strongly confounded by organ site, histologic subtype, and histomorphologic variables; therefore, it is important to provide situational context and instructions for clinical use of the CTA algorithms24,51,52. For example, a CTA algorithm designed for general-purpose breast cancer TILs scoring should be validated on different subtypes (infiltrating ductal, infiltrating lobular, mucinous, etc) and on a wide array of slides that capture variabilities in tumor phenotype (e.g., vacuolated tumor, necrotic tumor, etc), stromal phenotype (e.g., desmoplastic stroma), TIL densities, and sources of variability like staining and artifacts. That being said, it is plausible to assume that the biology and significance of TILs may vary in different clinical and genomic subtypes of the same primary cancer site, and that a general-purpose TILs-scoring algorithm may not be applicable. Further research into the commonalities and differences in the prognostic and biological value of TILs in different tissue sites and within different subtypes of the same cancer is warranted.

Clear inclusion criteria are helpful in deciding whether a slide is suitable for a particular CTA algorithm. For robust implementation, it is useful to: 1. detect when slides fail to meet its minimum quality; 2. provide some measure of confidence in its predictions; 3. be free of single points of failure (i.e., modular enough to tolerate failure of some sub-components); 4. be somewhat explainable, such that an expert pathologist can understand its limitations, common failure modes, and what the model seems to rely on in making decisions. Algorithms for measuring image quality and detecting artifacts will play an important role in the clinical implementation of CTA53.

From a computer vision perspective, we can subdivide CTA in two separate tasks: 1. segmentation of the region of interest (e.g., intratumoral stroma in case of sTIL assessment) and 2. detection of individual TILs within that region. In practice, a set of complementary computer vision problems often need to be addressed to score TILs (Fig. 1). To segment the region in which TILs will be assessed, it is also often needed to explicitly segment regions for exclusion from the analysis. Although these can be manually annotated by pathologists, these judgements are a significant source of variability in VTA, and developing algorithms capable of performing these tasks could improve reproducibility and standardization7,8,9.

Fig. 1: Outline of the visual (VTA) and computational (CTA) procedure for scoring TILs in breast carcinomas.

figure 1

TIL scoring is a complex procedure, and breast carcinomas are used as an example. Specific guidelines for scoring different tumors are provided in the references. Steps involved in VTA and/or CTA are tagged with these abbreviations. CTA according to TIL-WG guidelines involves TIL scoring in different tissue compartments. a Invasive edge is determined (red) and key confounding regions like necrosis (yellow) are delineated. b Within the central tumor, tumor-associated stroma is determined (green). Other considerations and steps are involved depending on histologic subtype, slide quality, and clinical context. c Determination of regions for inclusion or exclusion in the analysis in accordance with published guidelines. d Final score is estimated (visually) or calculated (computationally). In breast carcinomas, stromal TIL score (sTIL) is used clinically. Intratumoral TIL score (iTIL) is subject to more VTA variability, which has hampered the generation of evidence demonstrating prognostic value; perhaps CTA of iTILs will prove less variable and, consequently, prognostic. e The necessity of diverse pathologist annotations for robust analytical validation of computational models. Desmoplastic stroma may be misclassified as tumor regions; Vacuolated tumor may be misclassified as stroma; intermixed normal acini or ducts, DCIS/LCIS, and blood vessels may be misclassified as tumor; plasma cells are sometimes misclassified as carcinoma cells. Note that while the term “TILs” includes lymphocytes, plasma cells and other small mononuclear infiltrates, lumping these categories may not be optimal from an algorithm design perspective; plasma cells tend to be morphologically different from lymphocytes in nuclear texture, size, and visible cytoplasm. f Various computational approaches may be used for computational scoring. The more granular the algorithm is, the more accurate/useful it is likely to be, but—as a trade-off—the more it relies on exhaustive manual annotations from pathologists. The least granular approach is patch classification, followed by region delineation (segmentation), then object detection (individual TILs). A robust computational scoring algorithm likely utilizes a combination of these (and related) approaches.

Full size image

Specifically, segmentation of the “central tumor” and the “invasive margin/edge” enable TILs quantitation to be focused in relevant areas, excluding “distant” stroma along with normal tissue and surrounding structures. A semi-precise segmentation of invasive margin also allows sTILs score to be broken down for the margin and central tumor regions (especially, in colorectal carcinomas) and to characterize peri-tumoral TILs independently10. Within the central tumor, segmenting carcinoma cell nests and intratumoral stroma enables separate measurements for sTIL and iTIL densities. Furthermore, segmentation helps exclude key confounder regions that need to be excluded from the analysis. This includes necrosis, tertiary lymphoid structures, intermixed normal tissue or DCIS/LCIS (in breast carcinoma), pre-existing lymphoid stroma (in lymph nodes and oropharyngeal tumors), perivascular regions, intra-alveolar regions (in lung), artifacts, etc. This step requires high-quality segmentation annotations, and may prove to be challenging. Indeed, for routine clinical practice, it may be necessary to have a pathologist perform a quick visual confirmation of algorithmic region segmentations, and/or create high-level region annotations that may be difficult to produce algorithmically.

When designing a TIL classifier, consideration of key confounding cells is important. Although lymphocytes are, compared with tumor cells, relatively monomorphic, their small sizes offer little lymphocyte-specific texture information; small or perpendicularly cut stromal cells and even prominent nucleoli may result in misclassifications. Apoptotic bodies, necrotic debris, neutrophils, and some tumor cells (especially in lobular breast carcinomas and small-blue-round cell tumors) are other common confounders. Quantitation of systematic misclassification errors is warranted; some misclassifications will have contradictory consequences for clinical decision making. For example, neutrophils are evidently associated with adverse clinical outcomes, whereas TILs are typically associated with favorable outcomes51. Note that some of the TIL-WG clinical guidelines have been optimized for human scoring and are not very applicable in CTA algorithm design. For example, in breast carcinomas it is advised to “include but not focus on” tumor invasive edge TILs and TILs “hotspots”; CTA circumvents the need to address these cognitive biases11. To fully adhere to clinical guidelines, segmentation of TILs is warranted, so that the fraction of intratumoral stroma occupied by TILs is calculated.

Computer-aided versus fully automated TILs assessment

The extent to which computational tools can be used to complement clinical decision making is highly context-dependent, and is strongly impacted by cancer type and clinical setting54,55,56,57. In a computer-aided diagnosis paradigm, CTA is only used to provide guidance and increase efficiency in the workflow by any combination of the following: 1. calculating overall TILs score estimates to provide a frame-of-reference for the visual estimate; 2. directing the pathologist attention to regions of interest for TIL scoring, helping mitigate inconsistencies caused by heterogeneity in TILs density in different regions within the same slide; 3. providing a quantitative estimate for TILs density within regions of interest that the pathologist identifies, hence reducing ambiguity in visual estimation. Two models exist to assess this type of workflow during model development. In the traditional open assessment framework, the algorithm is trained on a set of manually annotated data points and evaluated on an independent held-out testing set. Alternatively, a closed-loop framework may be adopted, whereby pathologists can use the algorithmic output to re-evaluate their original decisions on the held-out set after exposure to the algorithmic results55,56. Both frameworks have pros and cons, although the closed-loop framework enables assessment of the potential impact that CTA has on altering the clinical decision-making process56.

The alternative paradigm is an entirely computational pipeline for CTA. This approach clearly provides efficiency gains, which could markedly reduce costs and accelerate development in a research setting. When the sample sizes are large enough, a few failures (i.e., “noise”) could be tolerated without altering the overall conclusions. This is contrary to clinical medicine, where CTA is expected to be highly dependable for each patient, especially when it is used to guide treatment decisions. Owing to the highly consequential nature of medical decision-making, a stand-alone CTA algorithm requires a higher bar for validation. It is also likely that even validated stand-alone CTA tools will need “sanity checks” by pathologists, guarding against unexpected failures. For example, a CTA report may be linked to a WSI display system to visualize the intermediate results (i.e., detected tissue boundaries and TILs locations) that were used by the algorithm to reach its decision (Fig. 2).

Fig. 2: Conceptual pathology report for computational TIL assessment (CTA).

figure 2

CTA reports might include global TIL estimates, broken down by key histologic regions, and estimates of classifier confidence. CTA reports are inseparably linked to WSI viewing systems, where algorithmic segmentations and localizations supporting the calculated scores are displayed for sanity check verification by the attending pathologist. Other elements, like local TIL estimates, TIL clustering results, and survival predictions may also be included.

Full size image

We do not envision computational models at their current level of performance replacing pathologist expertize. In fact, we would argue that quite the opposite is true; CTA enables objective quantitative assessment of an otherwise ambiguous metric, enabling the pathologist to focus more of his/her time on higher-order decision-making tasks54. With that in mind, we argue that the efficiency gains from CTA in under-resourced settings are likely to be derived from workflow efficiency, as opposed to reducing the domain expertize required to make diagnostic and therapeutic assessments. When used in a telepathology setting, i.e., off-site review of WSIs, CTA is still likely to require supervision by an experienced attending pathologist. Naturally, this depends on infrastructure, and one may argue that the cost-effectiveness of CTA is determined by the balance between infrastructure costs (WSI scanners, computing facilities, software, cloud support, etc) and expected long-term efficiency gains.

Validation and training issues surrounding computational TIL scoring

CTA algorithms will need to be validated just like any prognostic or predictive biomarker to demonstrate preanalytical validation (Pre-AV), analytical validation (AV), clinical validation (CV), and clinical utility8,58,59. In brief, Pre-AV is concerned with procedures that occur before CTA algorithms are applied, and include items like specimen preparation, slide quality, WSI scanner magnification and specifications, image format, etc; AV refers to accuracy and reproducibility; CV refers to stratification of patients into clinically meaningful subgroups; clinical utility refers to overall benefit in the clinical setting, considering existing methods and practices. Other considerations include cost-effectiveness, implementation feasibility, and ethical implications59. VTA has been subject to extensive AV, CV, and clinical utility assessment, and it is critical that CTA algorithms are validated using the same high standards7,8. The use-case of a CTA algorithm, specifically whether it is used for computer-aided assessment or for largely unsupervised assessment, is a key determinant of the extent of required validation. Key resources to consult include: 1. Recommendations by the Society for Immunotherapy of Cancer, for validation of diagnostic biomarkers; 2. Guidance documents by the US Food and Drug Administration (FDA); 3. Guidelines from the College of American Pathologists, for validation of diagnostic WSI systems60,61,62,[63](#ref-CR63 "Fda, M. Guidance for Industry and Food and Drug Administration Staff - Technical Performance Assessment of Digital Pathology Whole Slide Imaging Devices. (2016). Available at: https://www.fda.gov/media/90791/download

            ."),[64](/articles/s41523-020-0154-2#ref-CR64 "US Food and Drug Administration. Device Advice for AI and Machine Learning Algorithms. NCIPhub - Food and Drug Administration. Available at: 
              https://nciphub.org/groups/eedapstudies/wiki/DeviceAdvice
              
            . (Accessed: 4th July 2017)."). Granted, some of these require modifications in the CTA context, and we will highlight some of these differences here.

Pre-AV is of paramount importance, as CTA algorithm performance may vary in the presence of artifacts, variability in staining, tissue thickness, cutting angle, imaging, and storage65,66,67,68. Trained pathologists, on the other hand, are more agile in adapting to variations in tissue processing, although these factors can still impact their visual assessment. Some studies have shown that the implementation of a DICOM standard for pathology images can improve standardization and improve interoperability if adopted by manufacturers67,69. Techniques for making algorithms robust to variations, rather than eliminating the variations, have also been widely studied and are commonly employed69,70,[71](#ref-CR71 "Tellez, D. et al. Quantifying the effects of data augmentation and stain color normalization in convolutional neural networks for computational pathology. http://arxiv.org/abs/1902.06543

             (2019)."),[72](/articles/s41523-020-0154-2#ref-CR72 "Hou, L. et al. Unsupervised histopathology image synthesis. 
              http://arxiv.org/abs/1712.05021
              
             (2017)."). According to CAP guidelines, it is necessary to perform in-house validation of CTAs in all pathology laboratories, to validate the entire workflow (i.e., for each combination of tissue, stain, scanner, and CTA) using adequate sample size representing the entire diagnostic spectrum, and to re-validate whenever a significant component of the pre-analytic workflow changes[62](/articles/s41523-020-0154-2#ref-CR62 "Pantanowitz, L. et al. Validating whole slide imaging for diagnostic purposes in pathology: guideline from the College of American Pathologists Pathology and Laboratory Quality Center. Arch. Pathol. Lab. Med. 137, 1710–1722 (2013)."). Pre-AV and AV are most suitable in the in-house validation setting, as they can be performed with relatively fewer slides. It may be argued that proper in-house Pre-AV and AV suffice, provided large-scale prospective (or retrospective-prospective) AV, CV, and Clinical Utility studies were performed in a multi-center setting. Demonstrating local equivalency of Pre-AV and AV results can thus allow “linkage” to existing CV and Clinical Utility results assuming comparable patient populations.

AV typically involves quantitative assessment of CTA algorithm performance using ML metrics like segmentation or classification accuracy, prediction vs truth error, and area under receiver–operator characteristic curve or precision-recall curves. AV also includes validation against “non-classical” forms of ground truth like co-registered IHC, in which case the registration process itself may also require validation. AV is a necessary prerequisite to CV as it answers the more fundamental question: “Do CTA algorithms detect TILs correctly?”. AV should measure performance over the spectrum of variability induced by pre-analytic factors, and in cohorts that reflect the full range of intrinsic/biological variability. Naturally, this means that uncommon or rare subtypes of patterns are harder to validate owing to sample size limitations. AV of nucleus detection and classification algorithms has often neglected these issues, focusing on a large number of cells from a small number of cases.

Demonstrating the validity and generalization of prediction models is a complex process. Typically, the initial focus is on “internal” validation, using techniques like split-sample cross validation and bootstrapping. Later, the focus shifts to “external” validation, i.e., on an independent cohort from another institution. A hybrid technique called “internal–external” (cross-) validation may be appropriate when multi-institutional data sets (like the TCGA and METABRIC) are available, where training is performed on some hospitals/institutions and validation is performed on others. This was recommended by Steyerberg and Harrell and used in some computational pathology studies16,73,74,75.

Many of the events associated with cancer progression and subtyping are strongly correlated, so it may not be enough to show correspondence between global/slide-level CTA and VTA scores, as this shortcuts the AV process49. AV therefore relies on the presence of quality “ground truth” annotations. Unfortunately, there is a lack of open-access, large-scale, multi-institutional histology segmentation and/or TIL classification data sets, with few exceptions16,24,76,77. To help address this, a group of scientists, including the US FDA Center for Devices and Radiological Health (CDRH) and the TIL-WG, is collaborating to crowdsource pathologists and collect images and pathologist annotations that can be qualified by the FDA/CDRH medical device development tool program (MDDT). The MDDT qualified data would be available to any algorithm developer to be used for the analytic evaluation of their algorithm performance in a submission to the FDA/CDRH[78](/articles/s41523-020-0154-2#ref-CR78 "US Food and Drug Administration. Year 2: High-throughput truthing of microscope slides to validate artificial intelligence algorithms analyzing digital scans of pathology slides: data (images + annotations) as an FDA-qualified medical device development tool (MDDT). Available at: https://ncihub.org/groups/eedapstudies/wiki/HighthroughputTruthingYear2?version=2

            ."). The concept of “ground truth” in pathology can be vague and is often subjective, especially when dealing with H&E; it is therefore important to measure inter-rater variability by having multiple experts annotate the same regions and objects[7](/articles/s41523-020-0154-2#ref-CR7 "Denkert, C. et al. Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international immuno-oncology biomarker working group. Mod. Pathol. 29, 1155–1164 (2016)."),[8](/articles/s41523-020-0154-2#ref-CR8 "Wein, L. et al. Clinical validity and utility of tumor-infiltrating lymphocytes in routine clinical practice for breast cancer patients: current and future directions. Front. Oncol. 7, 156 (2017)."). A key bottleneck in this process is the time commitment of pathologists, so collaborative, educational and/or crowdsourcing settings can help circumvent this limitation[16](/articles/s41523-020-0154-2#ref-CR16 "Amgad, M. et al. Structured crowdsourcing enables convolutional segmentation of histology images. Bioinformatics 35, 3461–3467 (2019)."),[79](/articles/s41523-020-0154-2#ref-CR79 "Ørting, S. et al. A survey of crowdsourcing in medical image analysis. 
              http://arxiv.org/abs/1902.09159
              
             (2019)."). It should be stressed, however, that although annotations from non-pathologists or residents may be adequate for CTA algorithm training; validation may require ground truth annotations created or reviewed by experienced practicing pathologists[16](/articles/s41523-020-0154-2#ref-CR16 "Amgad, M. et al. Structured crowdsourcing enables convolutional segmentation of histology images. Bioinformatics 35, 3461–3467 (2019)."),[80](/articles/s41523-020-0154-2#ref-CR80 "Hannun, A. Y. et al. Cardiologist-level arrhythmia detection and classification in ambulatory electrocardiograms using a deep neural network. Nat. Med. 25, 65–69 (2019).").

It is important to note that the ambiguity in ground truth (even if determined by consensus by multiple pathologists) typically warrants additional validation using objective criteria, most notably the ability to predict concrete clinical endpoints in validated data sets. One of the best ways to meet this validation bar is to use WSIs from large, multi-institutional randomized-controlled trials. To facilitate this effort, the TIL-WG is establishing strategic international partnerships to organize a machine learning challenge to validate CTA algorithms using clinical trials data. The training sets would be made available for investigators to train and fine tune their models, whereas separate blinded validation sets would only be provided once a locked-down algorithm has been established. Such resources are needed so that different algorithms and approaches can be directly compared on the same, high-quality data sets.

CTA for clinical versus academic use

Like VTA, CTA may be considered to fall under the umbrella of “imaging biomarkers,” and likely follows a similar validation roadmap to enable clinical translation and adoption38,81,82. CTA may be used in the following academic settings, to name a few: 1. as a surrogate marker of response to experimental therapy in animal models; 2. as a diagnostic or predictive biomarker in retrospective clinical studies using archival WSI data; 3. as a diagnostic or predictive biomarker in prospective randomized-controlled trials. Incorporation of imaging biomarkers into prospective clinical trials requires some form of analytical and clinical validation (using retrospective data, for example), resulting in the establishment of Standards of Practice for trial use81. Establishment of clinical validity and utility in multicentric prospective trials is typically a prerequisite for use in day-to-day clinical practice. In a research environment, it is not unusual for computational algorithms to be frequently tweaked in a closed-loop fashion. This tweaking can be as simple as altering hyper-parameters, but can include more drastic changes like modifications to the algorithm or (inter)active machine learning83,84. From a standard regulatory perspective, this is problematic as validation requires a defined “lockdown” and version control; any change generally requires at least partial re-validation[64](/articles/s41523-020-0154-2#ref-CR64 "US Food and Drug Administration. Device Advice for AI and Machine Learning Algorithms. NCIPhub - Food and Drug Administration. Available at: https://nciphub.org/groups/eedapstudies/wiki/DeviceAdvice

            . (Accessed: 4th July 2017)."),[85](/articles/s41523-020-0154-2#ref-CR85 "U.S. Food and Drug Administration (FDA). Considerations for Design, Development, and Analytical Validation of Next Generation Sequencing-Based In Vitro Diagnostics Intended to Aim in the Diagnosis of Suspected Germline Diseases. (2018). Available at: 
              https://www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/UCM509838.pdf
              
            ."). It is therefore clear that the most pronounced difference between CTA use in basic/retrospective research, prospective trials, and routine clinical setting is the rigor of validation required[38](/articles/s41523-020-0154-2#ref-CR38 "Hamilton, P. W. et al. Digital pathology and image analysis in tissue biomarker research. Methods 70, 59–73 (2014)."),[81](/articles/s41523-020-0154-2#ref-CR81 "O’Connor, J. P. B. et al. Imaging biomarker roadmap for cancer studies. Nat. Rev. Clin. Oncol. 14, 169–186 (2017)."),[82](/articles/s41523-020-0154-2#ref-CR82 "Abramson, R. G. et al. Methods and challenges in quantitative imaging biomarker development. Acad. Radiol. 22, 25–32 (2015).").

In a basic/retrospective research environment, there is naturally a higher degree of flexibility in adopting CTA algorithms. For example, all slides may be scanned using the same scanner and using similar tissue processing protocols. In this setting, there is no immediate need for worrying about algorithm generalization performance under external processing or scanning conditions. Likewise, it may not be necessary to validate the model using ground truth from multiple pathologists, especially if some degree of noise can be tolerated. Operational issues and practicality also play a smaller role in basic/retrospective research settings; algorithm speed and user friendliness of a particular CTA algorithm may not be relevant when routine/repetitive TILs assessment is not needed. Even the nature of CTA algorithms may be different in a non-clinical setting. For instance, even though there is conflicting evidence on the prognostic value of iTILs in breast cancer, there are motivations to quantify them in a research environment. It should be noted, however, that this flexibility is only applicable for CTA algorithms that are being used to support non-clinical research projects, not for those algorithms that are being validated for future clinical use.

The future of computational image-based immune biomarkers

CTA algorithms can enable characterization of the tumor microenvironment beyond the limits of human observers, and will be an important tool in identifying latent prognostic and predictive patterns of immune response. For one, CTA enables calculation of local TIL densities at various scales, which may serve as a guide to “pockets” of differential immune activation (Fig. 2). This surpasses what is possible with VTA and such measurements are easy to calculate provided that CTA algorithms detect TILs with adequate sensitivity and specificity. Several studies have identified genomic features that in hindsight are associated with TILs, and CTA presents opportunities for systematic investigation of these associations24,26,74,86,87. The emergence of assays and imaging platforms for multiplexed immunofluorescence and in situ hybridization will present new horizons for identifying predictive immunologic patterns and for understanding the molecular basis of tumor-immune interactions88,89; these approaches are increasingly becoming commoditized.

Previous work examined how various spatial metrics from cancer-associated stroma relate to clinical outcomes, and similar concepts can be borrowed; for example, metrics capturing the complex relationships between TILs and other cells/structures in the tumor microenvironment90. CTA may enable precise definitions of “intratumoral stroma”, for example using a quantitative threshold (i.e., “stroma within x microns from nearest tumor nest”). Similar concepts could be applied when differentiating tertiary lymphocytic aggregates, or other TIL hotspots, from infiltrating TILs that presumably have a direct role in anticancer response. It is also important to note that lymphocytic aggregation and other higher-order quantitative spatial metrics may play important prognostic roles yet to be discovered. A CTA study identified five broad categories of spatial organization of TILs infiltration, which are differentially associated with different cancer sites and subtypes24. Alternatively, TILs can be placed on a continuum, such that sTILs that have a closer proximity to carcinoma nests get a higher weight. iTILs could be characterized using similar reasoning. Depending on available ground truth, numerous spatial metrics can be calculated. Nuanced assessment of immune response can be performed; for example, number of apoptotic bodies and their relation to nearby immune infiltrates. It is likely that there would be a considerable degree of redundancy in the prognostic value of CTA metrics; such redundancy is not uncommon in genomic biomarkers91. This should not be problematic as long as statistical models properly account for correlated predictors. In fact, the ability to calculate numerous metrics for a very large volume of cases enables large-scale, systematic discovery of histological biomarkers, bringing us a step closer to evidence-based pathology practice.

Learning-based algorithms can be utilized to learn prognostic features directly from images in a minimally biased manner (without explicit detection of TILs), and to integrate these with standard clinico-pathologic and genomic predictors. The approach of using deep learning algorithms to first detect and classify TILs and structures in histology, and then to calculate quantitative features of these objects, presents a way of closely modeling the clinical guidelines set forth by expert pathologists. Here, the power of learning algorithms is directed at providing highly accurate and robust detection and classification to enable reproducible and quantitative measurement. Although this approach is interpretable and provides a clear path for analytic validation, the limitation is that quantitative features are prescribed instead of learned. Recently, there have been successful efforts to develop end-to-end prognostic deep learning models that learn to directly predict clinical outcomes from raw images without any intermediate classification of histologic objects like TILs17,[92](/articles/s41523-020-0154-2#ref-CR92 "Meier, A. et al. 77PEnd-to-end learning to predict survival in patients with gastric cancer using convolutional neural networks. Ann. Oncol. 29 https://doi.org/10.1093/annonc/mdy269.07510.1093/annonc/mdy269.075

             (2018)."). Although these end-to-end learning approaches have the potential to learn latent prognostic patterns (including those impossible to assess visually), they are less interpretable and thus the factors driving the predictions are currently unknown.

Finally, we would note that one of the key limitations of machine learning models, and deep learning models in particular, is their opaqueness. It is often the case that model accuracy comes at a cost to explainability, giving rise to the term “black box” often associated with deep learning. The problem with less explainable models is that key features driving output may not be readily identifiable to evaluate biologic plausibility, and hence the only safeguard against major flaws is extensive validation93. Perhaps the most notorious consequence of this problem is “adversarial examples”, which are images that look natural to the human eye but that are specifically crafted (e.g., by malicious actors) to mislead deep learning models to make targeted misclassifications94. Nevertheless, recent advances in deep learning research have substantially increased model interpretability, and have devised key model training strategies (e.g., generative adversarial neural networks) to increase performance robustness93,95,96,97.

Conclusions

Advances in digital pathology and ML methodology have yielded expert-level performance in challenging diagnostic tasks. Evaluation of TILs in solid tumors is a highly suitable application for computational and computer-aided assessment, as it is both technically feasible and fills an unmet clinical need for objective and reproducible assessment. CTA algorithms need to account for the complexity involved in TIL-scoring procedures, and to closely follow guidelines for visual assessment where appropriate. TIL scoring needs to capture the concepts of stromal and intratumoral TILs and to account for confounding morphologies specific to different tumor sites, subtypes, and histologic patterns. Preanalytical factors related to imaging modality, staining procedure, and slide inclusion criteria are critical considerations, and robust analytical and clinical validation is key to adoption. In the clinical setting, CTA would ideally provide time- and cost-savings for pathologists, who face increasing demands for reporting biomarkers that are time-consuming to evaluate and subject to considerable inter- and intra- reader variability. In addition, CTA enables discovery of complex spatial patterns and genomic associations beyond the limits of visual scoring, and presents opportunities for precision medicine and scientific discovery.

References

  1. Piccart-Gebhart, M. et al. Adjuvant lapatinib and trastuzumab for early human epidermal growth factor receptor 2-positive breast cancer: results from the randomized phase III adjuvant lapatinib and/or Trastuzumab Treatment Optimization Trial. J. Clin. Oncol. 34, 1034–1042 (2016).
    Article CAS PubMed Google Scholar
  2. von Minckwitz, G. et al. Adjuvant pertuzumab and trastuzumab in early HER2-positive breast cancer. N. Engl. J. Med. 377, 122–131 (2017).
    Article Google Scholar
  3. Denkert, C. et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J. Clin. Oncol. 28, 105–113 (2010).
    Article CAS PubMed Google Scholar
  4. Savas, P. et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat. Rev. Clin. Oncol. 13, 228–241 (2016).
    Article CAS PubMed Google Scholar
  5. Burns, P. B., Rohrich, R. J. & Chung, K. C. The levels of evidence and their role in evidence-based medicine. Plast. Reconstr. Surg. 128, 305–310 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  6. Balic, M., Thomssen, C., Würstlein, R., Gnant, M. & Harbeck, N. St. Gallen/Vienna 2019: a brief summary of the consensus discussion on the optimal primary breast cancer treatment. Breast Care 14, 103–110 (2019).
  7. Denkert, C. et al. Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international immuno-oncology biomarker working group. Mod. Pathol. 29, 1155–1164 (2016).
    Article CAS PubMed Google Scholar
  8. Wein, L. et al. Clinical validity and utility of tumor-infiltrating lymphocytes in routine clinical practice for breast cancer patients: current and future directions. Front. Oncol. 7, 156 (2017).
    Article PubMed PubMed Central Google Scholar
  9. Brambilla, E. et al. Prognostic effect of tumor lymphocytic infiltration in resectable non-small-cell lung cancer. J. Clin. Oncol. 34, 1223–1230 (2016).
    Article CAS PubMed PubMed Central Google Scholar
  10. Hendry, S. et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international immunooncology biomarkers working group: part 1: assessing the host immune response, tils in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv. Anat. Pathol. 24, 235–251 (2017).
    Article PubMed PubMed Central Google Scholar
  11. Salgado, R. et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann. Oncol. 26, 259–271 (2015).
    Article CAS PubMed Google Scholar
  12. Hendry, S. et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international immuno-oncology biomarkers working group: part 2: tils in melanoma, gastrointestinal tract carcinomas, non-small cell lung carcinoma and mesothelioma, endometrial and ovarian carcinomas, squamous cell carcinoma of the head and neck, genitourinary carcinomas, and primary brain tumors. Adv. Anat. Pathol. 24, 311–335 (2017).
    Article PubMed PubMed Central Google Scholar
  13. Brunyé, T. T., Mercan, E., Weaver, D. L. & Elmore, J. G. Accuracy is in the eyes of the pathologist: the visual interpretive process and diagnostic accuracy with digital whole slide images. J. Biomed. Inform. 66, 171–179 (2017).
    Article PubMed PubMed Central Google Scholar
  14. Ehteshami Bejnordi, B. et al. Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA 318, 2199–2210 (2017).
    Article PubMed PubMed Central Google Scholar
  15. Tellez, D. et al. Whole-slide mitosis detection in H&E breast histology using PHH3 as a reference to train distilled stain-invariant convolutional networks. IEEE Trans. Med. Imaging (2018). https://doi.org/10.1109/TMI.2018.2820199
  16. Amgad, M. et al. Structured crowdsourcing enables convolutional segmentation of histology images. Bioinformatics 35, 3461–3467 (2019).
    Article CAS PubMed PubMed Central Google Scholar
  17. Mobadersany, P. et al. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc. Natl Acad. Sci. USA 115, E2970–E2979 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  18. Litjens, G. et al. A survey on deep learning in medical image analysis. Med. Image Anal. 42, 60–88 (2017).
    Article PubMed Google Scholar
  19. Janowczyk, A. & Madabhushi, A. Deep learning for digital pathology image analysis: a comprehensive tutorial with selected use cases. J. Pathol. Inform. 7, 29 (2016).
    Article PubMed PubMed Central Google Scholar
  20. Klauschen, F. et al. Scoring of tumor-infiltrating lymphocytes: from visual estimation to machine learning. Semin. Cancer Biol. 52, 151–157 (2018).
    Article CAS PubMed Google Scholar
  21. Barua, S. et al. Spatial interaction of tumor cells and regulatory T cells correlates with survival in non-small cell lung cancer. Lung Cancer 117, 73–79 (2018).
    Article PubMed Google Scholar
  22. Schalper, K. A. et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J. Natl. Cancer Inst. 107, dju435 (2015).
  23. Brown, J. R. et al. Multiplexed quantitative analysis of CD3, CD8, and CD20 predicts response to neoadjuvant chemotherapy in breast cancer. Clin. Cancer Res. 20, 5995–6005 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  24. Saltz, J. et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep. 23, 181–193.e7 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  25. Amgad, M. et al. Joint region and nucleus segmentation for characterization of tumor infiltrating lymphocytes in breast cancer. in Medical Imaging 2019: Digital Pathology (eds. Tomaszewski, J. E. & Ward, A. D.) 20 (SPIE, 2019).
  26. Yuan, Y. et al. Quantitative image analysis of cellular heterogeneity in breast tumors complements genomic profiling. Sci. Transl. Med. 4, 157ra143 (2012).
    Article PubMed Google Scholar
  27. Basavanhally, A. N. et al. Computerized image-based detection and grading of lymphocytic infiltration in HER2+ breast cancer histopathology. IEEE Trans. Biomed. Eng. 57, 642–653 (2010).
    Article PubMed Google Scholar
  28. Corredor, G. et al. Spatial architecture and arrangement of tumor-infiltrating lymphocytes for predicting likelihood of recurrence in early-stage non-small cell lung cancer. Clin. Cancer Res. 25, 1526–1534 (2019).
    Article CAS PubMed Google Scholar
  29. Yoon, H. H. et al. Intertumoral heterogeneity of CD3 and CD8 T-cell densities in the microenvironment of dna mismatch-repair-deficient colon cancers: implications for prognosis. Clin. Cancer Res. 25, 125–133 (2019).
    Article CAS PubMed Google Scholar
  30. Swiderska-Chadaj, Z. et al. Convolutional Neural Networks for Lymphocyte detection in Immunohistochemically Stained Whole-Slide Images. (2018).
  31. Heindl, A. et al. Relevance of spatial heterogeneity of immune infiltration for predicting risk of recurrence after endocrine therapy of ER+ breast cancer. J. Natl. Cancer Inst. 110, djx137 (2018).
  32. Stoler, M. H. Advances in cervical screening technology. Mod. Pathol. 13, 275–284 (2000).
    Article CAS PubMed Google Scholar
  33. Vis, J. Y. & Huisman, A. Verification and quality control of routine hematology analyzers. Int. J. Lab. Hematol. 38, 100–109 (2016).
    Article PubMed Google Scholar
  34. Perkel, J. M. Immunohistochemistry for the 21st century. Science 351, 1098–1100 (2016).
    Article Google Scholar
  35. Lloyd, M. C. et al. Using image analysis as a tool for assessment of prognostic and predictive biomarkers for breast cancer: how reliable is it? J. Pathol. Inform. 1, 29 (2010).
    Article PubMed PubMed Central Google Scholar
  36. Holten-Rossing, H., Møller Talman, M.-L., Kristensson, M. & Vainer, B. Optimizing HER2 assessment in breast cancer: application of automated image analysis. Breast Cancer Res. Treat. 152, 367–375 (2015).
    Article CAS PubMed Google Scholar
  37. Gavrielides, M. A., Gallas, B. D., Lenz, P., Badano, A. & Hewitt, S. M. Observer variability in the interpretation of HER2/neu immunohistochemical expression with unaided and computer-aided digital microscopy. Arch. Pathol. Lab. Med. 135, 233–242 (2011).
    Article PubMed PubMed Central Google Scholar
  38. Hamilton, P. W. et al. Digital pathology and image analysis in tissue biomarker research. Methods 70, 59–73 (2014).
    Article CAS PubMed Google Scholar
  39. Dieci, M. V. et al. Update on tumor-infiltrating lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in residual disease after neoadjuvant therapy and in carcinoma in situ: a report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. Semin. Cancer Biol. 52, 16–25 (2018).
    Article PubMed Google Scholar
  40. Finotello, F. & Trajanoski, Z. Quantifying tumor-infiltrating immune cells from transcriptomics data. Cancer Immunol. Immunother. 67, 1031–1040 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  41. Chakravarthy, A. et al. Pan-cancer deconvolution of tumour composition using DNA methylation. Nat. Commun. 9, 3220 (2018).
    Article PubMed PubMed Central CAS Google Scholar
  42. Ács, B. et al. Ki-67 as a controversial predictive and prognostic marker in breast cancer patients treated with neoadjuvant chemotherapy. Diagn. Pathol. 12, 20 (2017).
    Article PubMed PubMed Central CAS Google Scholar
  43. Yi, M. et al. Which threshold for ER positivity? a retrospective study based on 9639 patients. Ann. Oncol. 25, 1004–1011 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  44. Göranzon, C. et al. Immunohistochemical characterization of lymphocytes in microscopic colitis. J. Crohns. Colitis 7, e434–e442 (2013).
    Article PubMed Google Scholar
  45. Pagès, F. et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).
    Article PubMed Google Scholar
  46. Galon, J. et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J. Pathol. 232, 199–209 (2014).
    Article CAS PubMed Google Scholar
  47. Väyrynen, J. P. et al. An improved image analysis method for cell counting lends credibility to the prognostic significance of T cells in colorectal cancer. Virchows Arch. 460, 455–465 (2012).
    Article PubMed CAS Google Scholar
  48. Singh, U. et al. Analytical validation of quantitative immunohistochemical assays of tumor infiltrating lymphocyte biomarkers. Biotech. Histochem. 93, 411–423 (2018).
    Article CAS PubMed Google Scholar
  49. Buisseret, L. et al. Tumor-infiltrating lymphocyte composition, organization and PD-1/ PD-L1 expression are linked in breast cancer. Oncoimmunology 6, e1257452 (2017).
    Article PubMed CAS Google Scholar
  50. Blom, S. et al. Systems pathology by multiplexed immunohistochemistry and whole-slide digital image analysis. Sci. Rep. 7, 15580 (2017).
    Article PubMed PubMed Central CAS Google Scholar
  51. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).
    Article CAS PubMed PubMed Central Google Scholar
  52. Stanton, S. E., Adams, S. & Disis, M. L. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. JAMA Oncol. 2, 1354–1360 (2016).
    Article PubMed Google Scholar
  53. Janowczyk, A., Zuo, R., Gilmore, H., Feldman, M. & Madabhushi, A. HistoQC: an open-source quality control tool for digital pathology slides. JCO Clin. Cancer Inf. 3, 1–7 (2019).
    Google Scholar
  54. Hipp, J. et al. Computer aided diagnostic tools aim to empower rather than replace pathologists: Lessons learned from computational chess. J. Pathol. Inform. 2, 25 (2011).
    Article PubMed PubMed Central Google Scholar
  55. Gurcan, M. N. Histopathological image analysis: path to acceptance through evaluation. Microsc. Microanal. 22, 1004–1005 (2016).
    Article PubMed PubMed Central Google Scholar
  56. Fauzi, M. F. A. et al. Classification of follicular lymphoma: the effect of computer aid on pathologists grading. BMC Med. Inform. Decis. Mak. 15, 115 (2015).
    Article PubMed PubMed Central Google Scholar
  57. Madabhushi, A., Agner, S., Basavanhally, A., Doyle, S. & Lee, G. Computer-aided prognosis: predicting patient and disease outcome via quantitative fusion of multi-scale, multi-modal data. Comput. Med. Imaging Graph. 35, 506–514 (2011).
    Article PubMed Google Scholar
  58. Hayes, D. F. Precision medicine and testing for tumor biomarkers-are all tests born equal? JAMA Oncol. 4, 773–774 (2018).
    Article PubMed Google Scholar
  59. Selleck, M. J., Senthil, M. & Wall, N. R. Making meaningful clinical use of biomarkers. Biomark. Insights 12, 11772719–17715236 (2017).
  60. Masucci, G. V. et al. Validation of biomarkers to predict response to immunotherapy in cancer: Volume I - pre-analytical and analytical validation. J. Immunother. Cancer 4, 76 (2016).
    Article PubMed PubMed Central Google Scholar
  61. Dobbin, K. K. et al. Validation of biomarkers to predict response to immunotherapy in cancer: volume II - clinical validation and regulatory considerations. J. Immunother. Cancer 4, 77 (2016).
    Article PubMed PubMed Central Google Scholar
  62. Pantanowitz, L. et al. Validating whole slide imaging for diagnostic purposes in pathology: guideline from the College of American Pathologists Pathology and Laboratory Quality Center. Arch. Pathol. Lab. Med. 137, 1710–1722 (2013).
    Article PubMed PubMed Central Google Scholar
  63. Fda, M. Guidance for Industry and Food and Drug Administration Staff - Technical Performance Assessment of Digital Pathology Whole Slide Imaging Devices. (2016). Available at: https://www.fda.gov/media/90791/download.
  64. US Food and Drug Administration. Device Advice for AI and Machine Learning Algorithms. NCIPhub - Food and Drug Administration. Available at: https://nciphub.org/groups/eedapstudies/wiki/DeviceAdvice. (Accessed: 4th July 2017).
  65. Leo, P. et al. Evaluating stability of histomorphometric features across scanner and staining variations: prostate cancer diagnosis from whole slide images. J. Med. Imaging (Bellingham) 3, 047502 (2016).
    Article Google Scholar
  66. Pantanowitz, L., Liu, C., Huang, Y., Guo, H. & Rohde, G. K. Impact of altering various image parameters on human epidermal growth factor receptor 2 image analysis data quality. J. Pathol. Inform. 8, 39 (2017).
    Article PubMed PubMed Central Google Scholar
  67. Pantanowitz, L. et al. Twenty years of digital pathology: an overview of the road travelled, what is on the horizon, and the emergence of vendor-neutral archives. J. Pathol. Inform. 9, 40 (2018).
    Article PubMed PubMed Central Google Scholar
  68. Zarella, M. D., Yeoh, C., Breen, D. E. & Garcia, F. U. An alternative reference space for H&E color normalization. PLoS ONE 12, e0174489 (2017).
    Article PubMed PubMed Central CAS Google Scholar
  69. Herrmann, M. D. et al. Implementing the DICOM standard for digital pathology. J. Pathol. Inform. 9, 37 (2018).
    Article PubMed PubMed Central Google Scholar
  70. Van Eycke, Y.-R., Allard, J., Salmon, I., Debeir, O. & Decaestecker, C. Image processing in digital pathology: an opportunity to solve inter-batch variability of immunohistochemical staining. Sci. Rep. 7, 42964 (2017).
  71. Tellez, D. et al. Quantifying the effects of data augmentation and stain color normalization in convolutional neural networks for computational pathology. http://arxiv.org/abs/1902.06543 (2019).
  72. Hou, L. et al. Unsupervised histopathology image synthesis. http://arxiv.org/abs/1712.05021 (2017).
  73. Steyerberg, E. W. & Harrell, F. E. Jr. Prediction models need appropriate internal, internal-external, and external validation. J. Clin. Epidemiol. 69, 245–247 (2016).
    Article PubMed Google Scholar
  74. Natrajan, R. et al. Microenvironmental heterogeneity parallels breast cancer progression: a histology-genomic integration analysis. PLoS Med. 13, e1001961 (2016).
    Article PubMed PubMed Central Google Scholar
  75. Loi, S. et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast canc ers. J. Clin. Oncol. 37, 559–569 (2019).
    Article PubMed PubMed Central Google Scholar
  76. Beck, A. H. Open access to large scale datasets is needed to translate knowledge of cancer heterogeneity into better patient outcomes. PLoS Med. 12, e1001794 (2015).
    Article PubMed PubMed Central Google Scholar
  77. Litjens, G. et al. 1399 H&E-stained sentinel lymph node sections of breast cancer patients: the CAMELYON dataset. Gigascience 7, giy065 (2018).
  78. US Food and Drug Administration. Year 2: High-throughput truthing of microscope slides to validate artificial intelligence algorithms analyzing digital scans of pathology slides: data (images + annotations) as an FDA-qualified medical device development tool (MDDT). Available at: https://ncihub.org/groups/eedapstudies/wiki/HighthroughputTruthingYear2?version=2.
  79. Ørting, S. et al. A survey of crowdsourcing in medical image analysis. http://arxiv.org/abs/1902.09159 (2019).
  80. Hannun, A. Y. et al. Cardiologist-level arrhythmia detection and classification in ambulatory electrocardiograms using a deep neural network. Nat. Med. 25, 65–69 (2019).
    Article CAS PubMed PubMed Central Google Scholar
  81. O’Connor, J. P. B. et al. Imaging biomarker roadmap for cancer studies. Nat. Rev. Clin. Oncol. 14, 169–186 (2017).
    Article PubMed CAS Google Scholar
  82. Abramson, R. G. et al. Methods and challenges in quantitative imaging biomarker development. Acad. Radiol. 22, 25–32 (2015).
    Article PubMed PubMed Central Google Scholar
  83. Nalisnik, M. et al. Interactive phenotyping of large-scale histology imaging data with HistomicsML. Sci. Rep. 7, 14588 (2017).
    Article PubMed PubMed Central CAS Google Scholar
  84. Kwolek, B. et al. Breast Cancer Classification on Histopathological Images Affected by Data Imbalance Using Active Learning and Deep Convolutional Neural Network: 28th International Conference on Artificial Neural Networks, Munich, Germany, September 17–19, 2019, Proceedings. in Artificial Neural Networks and Machine Learning—ICANN 2019: Workshop and Special Sessions (eds. Tetko, I. V., Kůrková, V., Karpov, P. & Theis, F.) 11731, 299–312 (Springer International Publishing, 2019).
  85. U.S. Food and Drug Administration (FDA). Considerations for Design, Development, and Analytical Validation of Next Generation Sequencing-Based In Vitro Diagnostics Intended to Aim in the Diagnosis of Suspected Germline Diseases. (2018). Available at: https://www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/UCM509838.pdf.
  86. Mukherjee, A. et al. Associations between genomic stratification of breast cancer and centrally reviewed tumour pathology in the METABRIC cohort. NPJ Breast Cancer 4, 5 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  87. Cooper, L. A. et al. PanCancer insights from The Cancer Genome Atlas: the pathologist’s perspective. J. Pathol. 244, 512–524 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  88. Anderson, R. Multiplex fluorescence in situ hybridization (M-FISH). Methods Mol. Biol. 659, 83–97 (2010).
    Article CAS PubMed Google Scholar
  89. Longuespée, R. et al. Tissue proteomics for the next decade? Towards a molecular dimension in histology. OMICS 18, 539–552 (2014).
    Article PubMed CAS Google Scholar
  90. Beck, A. H. et al. Systematic analysis of breast cancer morphology uncovers stromal features associated with survival. Sci. Transl. Med. 3, 108ra113 (2011).
    Article PubMed Google Scholar
  91. Cantini, L. et al. Classification of gene signatures for their information value and functional redundancy. NPJ Syst. Biol. Appl 4, 2 (2018).
    Article PubMed Google Scholar
  92. Meier, A. et al. 77PEnd-to-end learning to predict survival in patients with gastric cancer using convolutional neural networks. Ann. Oncol. 29 https://doi.org/10.1093/annonc/mdy269.07510.1093/annonc/mdy269.075 (2018).
  93. Guidotti, R. et al. A survey of methods for explaining black box models. ACM Comput. Surv. 51, 1–42 (2018).
    Article Google Scholar
  94. Yuan, X., He, P., Zhu, Q. & Li, X. Adversarial examples: attacks and defenses for deep learning. IEEE Trans. Neural Netw. Learn Syst. 30, 2805–2824 (2019).
    Article PubMed Google Scholar
  95. Zhang, Q.-S. & Zhu, S.-C. Visual interpretability for deep learning: a survey. Front. Inf. Technol. Electron. Eng. 19, 27–39 (2018).
    Article Google Scholar
  96. Yousefi, S. et al. Predicting clinical outcomes from large scale cancer genomic profiles with deep survival models. Sci. Rep. 7, 11707 (2017).
    Article PubMed PubMed Central CAS Google Scholar
  97. Yi, X., Walia, E. & Babyn, P. Generative adversarial network in medical imaging: a review. Med. Image Anal. 58, 101552 (2019).
    Article PubMed Google Scholar

Download references

Acknowledgements

L.A.D.C. is supported in part by the National Institutes of Health National Cancer Institute (NCI) grants U01CA220401 and U24CA19436201. R.S. is supported by the Breast Cancer Research Foundation (BCRF), grant No. 17-194. J.S. is supported in part by NCI grants UG3CA225021 and U24CA215109. A.M. is supported in part by NCI grants 1U24CA199374-01, R01CA202752-01A1, R01CA208236-01A1, R01 CA216579-01A1, R01 CA220581-01A1, 1U01 CA239055-01, National Center for Research Resources under award number 1 C06 RR12463-01, VA Merit Review Award IBX004121A from the United States Department of Veterans Affairs Biomedical Laboratory Research and Development Service, the DOD Prostate Cancer Idea Development Award (W81XWH-15-1-0558), the DOD Lung Cancer Investigator-Initiated Translational Research Award (W81XWH-18-1-0440), the DOD Peer Reviewed Cancer Research Program (W81XWH-16-1-0329), the Ohio Third Frontier Technology Validation Fund, the Wallace H. Coulter Foundation Program in the Department of Biomedical Engineering and the Clinical and Translational Science Award Program (CTSA) at Case Western Reserve University. S.G. is supported by Susan G Komen Foundation (CCR CCR18547966) and a Young Investigator Grant from the Breast Cancer Alliance. T.O.N. receives funding support from the Canadian Cancer Society. M.M.S. is supported by P30 CA16672 DHHS/NCI Cancer Center Support Grant (CCSG). A.S. is supported in part by NCI grants 1UG3CA225021, 1U24CA215109, and Leidos 14 × 138. This work includes contributions from, and was reviewed by, individuals at the F.D.A. This work has been approved for publication by the agency, but it does not necessarily reflect official agency policy. Certain commercial materials and equipment are identified in order to adequately specify experimental procedures. In no case does such identification imply recommendation or endorsement by the FDA, nor does it imply that the items identified are necessarily the best available for the purpose. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health, the US Department of Veterans Affairs, the Department of Defense, the United States Government, or other governments or entities. The following is a list of current members of the International Immuno-Oncology Working Group (TILs Working Group). Members contributed to the manuscript through discussions, including at the yearly TIL-WG meeting, and have reviewed and provided input on the manuscript. The authors alone are responsible for the views expressed in the work of the TILs Working Group and they do not necessarily represent the decisions, policy or views of their employer.

Author information

Authors and Affiliations

  1. Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
    Mohamed Amgad & Ashish Sharma
  2. Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
    Elisabeth Specht Stovgaard & Eva Balslev
  3. DTU Compute, Department of Applied Mathematics, Technical University of Denmark, Lyngby, Denmark
    Jeppe Thagaard
  4. Visiopharm A/S, Hørsholm, Denmark
    Jeppe Thagaard
  5. FDA/CDRH/OSEL/Division of Imaging, Diagnostics, and Software Reliability, Silver Spring, MD, USA
    Weijie Chen, Sarah Dudgeon & Brandon D. Gallas
  6. PathAI, Cambridge, MA, USA
    Jennifer K. Kerner & Andrew H. Beck
  7. Institut für Pathologie, Universitätsklinikum Gießen und Marburg GmbH, Standort Marburg, Philipps-Universität Marburg, Marburg, Germany
    Carsten Denkert & Stephan Wienert
  8. Institute of Pathology, Philipps-University Marburg, Marburg, Germany
    Carsten Denkert
  9. German Cancer Consortium (DKTK), Partner Site Charité, Berlin, Germany
    Carsten Denkert
  10. Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
    Yinyin Yuan & Khalid AbdulJabbar
  11. Division of Molecular Pathology, The Institute of Cancer Research, London, UK
    Yinyin Yuan & Khalid AbdulJabbar
  12. Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Victoria, Australia
    Peter Savas, Roberto Salgado & Stephen J. Luen
  13. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
    Peter Savas, Sherene Loi & Prudence A. Francis
  14. Department of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Leonie Voorwerk
  15. Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH, USA
    Anant Madabhushi
  16. Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
    Anant Madabhushi
  17. Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Solna, Sweden
    Johan Hartman
  18. Departments of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Manu M. Sebastian
  19. Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Hugo M. Horlings
  20. Department of Research IT, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Jan Hudeček
  21. Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
    Francesco Ciompi & Jeroen A. W. M. van der Laak
  22. Department of Pathology, UCL Cancer Institute, London, UK
    David A. Moore
  23. Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
    Rajendra Singh
  24. Université Paris-Saclay, Univ. Paris-Sud, Villejuif, France
    Elvire Roblin
  25. Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
    Marcelo Luiz Balancin
  26. Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
    Marie-Christine Mathieu
  27. Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
    Jochen K. Lennerz
  28. Department of Histopathology, Manipal Hospitals Dwarka, New Delhi, India
    Pawan Kirtani
  29. Department of Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
    I-Chun Chen
  30. Nuffield Department of Population Health, University of Oxford, Oxford, UK
    Jeremy P. Braybrooke
  31. Department of Medical Oncology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
    Jeremy P. Braybrooke
  32. Pathology Department, Fondazione IRCCS Istituto Nazionale Tumori and University of Milan, School of Medicine, Milan, Italy
    Giancarlo Pruneri
  33. Weill Cornell Medical College, New York, NY, USA
    Sandra Demaria
  34. Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, USA
    Sylvia Adams
  35. Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
    Stuart J. Schnitt
  36. The University of Queensland Centre for Clinical Research and Pathology Queensland, Brisbane, Australia
    Sunil R. Lakhani
  37. Pathology Department, CIBERONC-Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
    Federico Rojo & Laura Comerma
  38. GEICAM-Spanish Breast Cancer Research Group, Madrid, Spain
    Federico Rojo & Laura Comerma
  39. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
    Sunil S. Badve
  40. Roche Tissue Diagnostics, Digital Pathology, Santa Clara, CA, USA
    Mehrnoush Khojasteh & Uday Kurkure
  41. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    W. Fraser Symmans & Alexander J. Lazar
  42. Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
    Christos Sotiriou
  43. ULB-Cancer Research Center (U-CRC) Université Libre de Bruxelles, Brussels, Belgium
    Christos Sotiriou
  44. Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
    Paula Gonzalez-Ericsson
  45. NRG Oncology/NSABP, Pittsburgh, PA, USA
    Katherine L. Pogue-Geile & Rim S. Kim
  46. Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
    David L. Rimm & Emily Reisenbichler
  47. Department of Pathology, IEO, European Institute of Oncology IRCCS & State University of Milan, Milan, Italy
    Giuseppe Viale
  48. Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Stephen M. Hewitt
  49. Ontario Institute for Cancer Research, Toronto, ON, Canada
    John M. S. Bartlett
  50. Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
    John M. S. Bartlett
  51. Department of Pathology and Molecular Pathology, Centre Jean Perrin, Clermont-Ferrand, France
    Frédérique Penault-Llorca
  52. UMR INSERM 1240, Universite Clermont Auvergne, Clermont-Ferrand, France
    Frédérique Penault-Llorca
  53. Victorian Comprehensive Cancer Centre building, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
    Shom Goel
  54. Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
    Huang-Chun Lien
  55. German Breast Group, c/o GBG-Forschungs GmbH, Neu-Isenburg, Germany
    Sibylle Loibl
  56. Department of Pathology, BC Cancer, Vancouver, British Columbia, Canada
    Zuzana Kos
  57. Peter MacCallum Cancer Centre, Melbourne, Australia
    Sherene Loi & Prudence A. Francis
  58. Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Matthew G. Hanna, Edi Brogi, Jorge Reis-Filho & Timothy d’Alfons
  59. Gustave Roussy, Universite Paris-Saclay, Villejuif, France
    Stefan Michiels & Damien Drubay
  60. Université Paris-Sud, Institut National de la Santé et de la Recherche Médicale, Villejuif, France
    Stefan Michiels & Damien Drubay
  61. Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Marleen Kok
  62. Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Marleen Kok
  63. University of British Columbia, Vancouver, British Columbia, Canada
    Torsten O. Nielsen & Sam Leung
  64. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Alexander J. Lazar
  65. Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Alexander J. Lazar
  66. Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Alexander J. Lazar
  67. Department of Pathology, Medical University of Vienna, Vienna, Austria
    Zsuzsanna Bago-Horvath
  68. GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
    Loes F. S. Kooreman
  69. Department of Pathology, Maastricht University Medical Centre, Maastricht, The Netherlands
    Loes F. S. Kooreman
  70. Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
    Jeroen A. W. M. van der Laak
  71. Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
    Joel Saltz
  72. Roche Diagnostics Information Solutions, Belmont, CA, USA
    Michael Barnes
  73. Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
    Roberto Salgado
  74. Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
    Lee A. D. Cooper
  75. Department of Oral and Maxillofacial Diseases, Helsinki, Finland
    Aini Hyytiäinen
  76. Department of Pathology, Matsuyama Shimin Hospital, Matsuyama, Japan
    Akira I. Hida
  77. Surgical Oncology, Baylor College of Medicine, Texas, USA
    Alastair Thompson
  78. Roche Diagnostics, Machelen, Belgium
    Alex Lefevre & Carolien Boeckx
  79. PhenoPath Laboratories, Seattle, USA
    Allen Gown
  80. Research Pathology, Genentech Inc., South San Francisco, USA
    Amy Lo, Harmut Koeppen, James Ziai & Jennifer Giltnane
  81. Department of Medical Sciences, University of Turin, Italy and Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
    Anna Sapino
  82. Pulmonary Pathology, New York University Center for Biospecimen Research and Development, New York University, New York, NY, USA
    Andre Moreira
  83. Department of Pathology, Johns Hopkins Hospital, Baltimore, USA
    Andrea Richardson
  84. Department of Pathology, Istituto Europeo di Oncologia, University of Milan, Milan, Italy
    Andrea Vingiani
  85. Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, USA
    Andrew M. Bellizzi
  86. Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
    Andrew Tutt
  87. Department of Oncology, Instituto Valenciano de Oncología, Valencia, Spain
    Angel Guerrero-Zotano
  88. Cancer Bioinformatics Lab, Cancer Centre at Guy’s Hospital, London, UK
    Anita Grigoriadis
  89. School of Life Sciences and Medicine, King’s College London, London, UK
    Anita Grigoriadis
  90. Department of Clinical Genetics and Pathology, Skåne University Hospital, Lund University, Lund, Sweden
    Anna Ehinger
  91. Dana-Farber Cancer Institute, Boston, MA, USA
    Anna C. Garrido-Castro
  92. Institut Curie, Paris Sciences Lettres Université, Inserm U934, Department of Pathology, Paris, France
    Anne Vincent-Salomon
  93. Department of Surgical Pathology Zealand University Hospital, Køge, Denmark
    Anne-Vibeke Laenkholm
  94. Departments of Pathology and Oncology, The Johns Hopkins Hospital, Baltimore, USA
    Ashley Cimino-Mathews
  95. National Surgical Adjuvant Breast and Bowel Project Operations Center/NRG Oncology, Pittsburgh, PA, USA
    Ashok Srinivasan & Soonmyung Paik
  96. Department of Pathology, Karolinska Institute, Solna, Sweden
    Balazs Acs
  97. Department of Pathology, New York University Langone Medical Centre, New York, USA
    Baljit Singh
  98. Department of Pathology and Laboratory Medicine, UNC School of Medicine, Columbia, USA
    Benjamin Calhoun
  99. Québec Heart and Lung Institute Research Center, Laval University, Quebec city, Quebec, Canada
    Benjamin Haibe-Kans & Venkata Manem
  100. Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
    Benjamin Solomon & Elizabeth F. Blackley
  101. Department of Medicine, University of Melbourne, Parkville, Australia
    Bibhusal Thapa
  102. Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
    Brad H. Nelson
  103. Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima, Perú
    Carlos Castaneda
  104. Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima, 15038, Peru
    Carlos Castaneda, Joselyn Sanchez & Miluska Castillo
  105. Providence Cancer Research Center, Portland, Oregon, USA
    Carmen Ballesteroes-Merino
  106. Department of Medical Oncology, Istituto Europeo di Oncologia, Milan, Italy
    Carmen Criscitiello
  107. Department of Pathology, AZ Turnhout, Turnhout, Belgium
    Cecile Colpaert
  108. Department of Pathology, St Vincent’s University Hospital and University College Dublin, Dublin, Ireland
    Cecily Quinn
  109. Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, USA
    Chakra S. Chennubhotla
  110. Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK
    Charles Swanton, Crispin Hiley & Maise al Bakir
  111. Azienda AUSL, Regional Hospital of Aosta, Aosta, Italy
    Cinzia Solinas
  112. University Hospital Halle (Saale), Institute of Pathology, Halle, Saale, Germany
    Daniel Bethmann
  113. Department of Pathology, Brigham and Women’s Hospital, Boston, MA Department of Pathology, Dana Farber Cancer Institute, Boston, MA, USA
    Deborah A. Dillon
  114. Department of Pathology, Jules Bordet Institute, Brussels, Belgium
    Denis Larsimont
  115. Department of Clinical Medicine, Macquarie University, Sydney, Australia
    Dhanusha Sabanathan
  116. HistoGeneX NV, Antwerp, Belgium and AZ Sint-Maarten Hospital, Mechelen, Belgium
    Dieter Peeters
  117. Oncology Clinical Development, Bristol-Myers Squibb, Princeton, USA
    Dimitrios Zardavas
  118. Institut für Pathologie, UK Hamburg, Hamburg, Germany
    Doris Höflmayer
  119. Department of Medicine, Vanderbilt University Medical Centre, Nashville, USA
    Douglas B. Johnson
  120. Department of Cancer Biology, Mayo Clinic, Jacksonville, USA
    E. Aubrey Thompson
  121. Department of Oncology, Mayo Clinic, Rochester, USA
    Edith Perez, Matthew P. Goetz & Roberto Leon-Ferre
  122. Roche, Tucson, USA
    Ehab A. ElGabry
  123. Department of Pathology, Universidad de La Frontera, Temuco, Chile
    Enrique Bellolio & Juan Carlos Araya
  124. Departamento de Anatomía Patológica, Universidad de La Frontera, Temuco, Chile
    Enrique Bellolio
  125. Tumor Pathology Department, Maria Sklodowska-Curie Memorial Cancer Center, Gliwice, Poland
    Ewa Chmielik
  126. Pathology and Tissue Analytics, Roche, Machelen, Belgium
    Fabien Gaire & Oliver Grimm
  127. Department of Medical Oncology, Gustave Roussy, Villejuif, France
    Fabrice Andre
  128. Sunnybrook Health Sciences Centre, Toronto, Canada
    Fang-I Lu
  129. Tehran University of Medical Sciences, Tehran, Iran
    Farid Azmoudeh-Ardalan
  130. Translational Research, Montreal, Canada
    Forbius Tina Gruosso
  131. Oncology Biomarker Development, Genentech-Roche, Machelen, Belgium
    Franklin Peale & Luciana Molinero
  132. Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, USA
    Fred R. Hirsch
  133. Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
    Frederick Klaushen
  134. Department of Pathology, Hospital de Oncología Maria Curie, Buenos Aires, Argentina
    Gabriela Acosta-Haab
  135. Directorate of Surgical Pathology, SA Pathology, Adelaide, Australia
    Gelareh Farshid
  136. Department of Pathology, GZA-ZNA Hospitals, Wilrijk, Belgium
    Gert van den Eynden
  137. University of Milano, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
    Giuseppe Curigliano
  138. Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS, Milan, Italy
    Giuseppe Curigliano
  139. Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, Leuven, Belgium
    Giuseppe Floris
  140. KU Leuven- University Hospitals Leuven, Department of Pathology, Leuven, Belgium
    Giuseppe Floris
  141. Department of Pathology, University Hospital Antwerp, Antwerp, Belgium
    Glenn Broeckx
  142. Translational Health Sciences, Department of Cellular Pathology, North Bristol NHS Trust, University of Bristol, Bristol, UK
    Harry R. Haynes
  143. Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, USA
    Heather McArthur
  144. Helsinki University Central Hospital, Helsinki, Finland
    Heikki Joensuu
  145. Department of Clinical Pathology, Akademiska University Hospital, Uppsala, Sweden
    Helena Olofsson
  146. International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France
    Ian Cree
  147. Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Iris Nederlof
  148. Department of Pathology, Sanatorio Mater Dei, Buenos Aires, Argentina
    Isabel Frahm
  149. Institute of Pathology, Medical University of Graz, Graz, Austria
    Iva Brcic
  150. Department of Oncology, National Cancer Centre, Singapore, Singapore
    Jack Chan
  151. Vivactiv Ltd, Bellingdon, Bucks, UK
    Jacqueline A. Hall
  152. Department of Pathology, Brigham and Women’s Hospital, Boston, USA
    Jane Brock
  153. Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
    Jelle Wesseling
  154. R&D UNICANCER, Paris, France
    Jerome Lemonnier
  155. Translational Sciences, MedImmune, Gaithersberg, USA
    Jiping Zha, Keith E. Steele & Marlon C. Rebelatto
  156. Breast Unit, Champalimaud Clinical Centre, Lisboa, Portugal
    Joana M. Ribeiro
  157. Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, USA
    Jodi M. Carter
  158. Department of Medicine, Clinical Division of Oncology, Comprehensive Cancer Centre Vienna, Medical University of Vienna, Vienna, Austria
    Johannes Hainfellner & Matthias Preusser
  159. Leicester Cancer Research Centre, University of Leicester, Leicester, and MRC Toxicology Unit, University of Cambridge, Cambridge, UK
    John Le Quesne
  160. Merck & Co., Inc, Kenilworth, NJ, USA
    Jonathan W. Juco, Kenneth Emancipator & Petar Jelinic
  161. Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Jorge Reis-Filho
  162. Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Jose van den Berg
  163. Department of Medicine, Department of Obstetrics & Gynecology and Women’s Health, Albert Einstein Medical Center, Bronx, USA
    Joseph Sparano
  164. GHI Le Raincy-Montfermeil, Chelles, Île-de-France, France
    Joël Cucherousset
  165. Department of Pathology, Gustave Roussy, Grand Paris, France
    Julien Adam
  166. Departments of Medicine and Cancer Biology, Vanderbilt University Medical Centre, Nashville, USA
    Justin M. Balko
  167. Vm Scope, Berlin, Germany
    Kai Saeger
  168. Department of Pathology, Breast Pathology Section, Northwestern University, Chicago, USA
    Kalliopi Siziopikou
  169. Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
    Karen Willard-Gallo & Laurence Buisseret
  170. Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Karolina Sikorska
  171. German Breast Group, Neu-Isenburg, Germany
    Karsten Weber
  172. Cancer Biomarkers Working Group, Faculty of Medicine and Pharmacy, Université Mohamed Premier, Oujda, Morocco
    Khalid El Bairi
  173. Yale Cancer Center Genetics, Genomics and Epigenetics Program, Yale School of Medicine, New Haven, CT, USA
    Kim R. M. Blenman & Lajos Pusztai
  174. Pathology Department, Stanford University Medical Centre, Stanford, USA
    Kimberly H. Allison
  175. Department of Pathology, University Hospital Ghent, Ghent, Belgium
    Koen K. van de Vijver
  176. Pathology and Tissue Analytics, Roche Innovation Centre Munich, Penzberg, Germany
    Konstanty Korski
  177. Center for Pharmacogenomics and Fudan-Zhangjiang, Center for Clinical Genomics School of Life Sciences and Shanghai Cancer Center, Fudan University, Fudan, China
    Leming Shi
  178. Sichuan Cancer Hospital, Chengdu, China
    Liu Shi-wei
  179. Biorepository and Tissue Technology Shared Resources, University of California San Diego, San Diego, USA
    M. Valeria Estrada
  180. Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
    Maartje van Seijen & Michiel de Maaker
  181. Department of Pathology, Gustave Roussy, Villejuif, France
    Magali Lacroix-Triki
  182. Institute of Cancer Research Clinical Trials and Statistics Unit, The Institute of Cancer Research, Surrey, UK
    Maggie C. U. Cheang
  183. Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
    Marc van de Vijver
  184. Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
    Maria Vittoria Dieci
  185. Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
    Martine Piccart
  186. Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Centre, Nashville, USA
    Melinda E. Sanders
  187. Harvard Medical School, Boston, USA
    Meredith M. Regan
  188. Division of Biostatistics, Dana-Farber Cancer Institute, Boston, USA
    Meredith M. Regan
  189. Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Australia
    Michael Christie
  190. Vernon Cancer Center, Newton-Wellesley Hospital, Newton, USA
    Michael Misialek
  191. Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
    Michail Ignatiadis
  192. Department of Pathology, Cliniques universitaires Saint-Luc, Brussels, Belgium
    Mieke van Bockstal
  193. Breast Center, Dept. OB&GYN and CCC (LMU), University of Munich, Munich, Germany
    Nadia Harbeck
  194. Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, USA
    Nadine Tung
  195. University of Leuven, Leuven, Belgium
    Nele Laudus
  196. Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
    Nicolas Sirtaine & Roland de Wind
  197. German Breast Group GmbH, Neu-Isenburg, Germany
    Nicole Burchardi
  198. Service de Biostatistique et d’Epidémiologie, Gustave Roussy, CESP, Université-Paris Sud, Université Paris-Saclay, Villejuif, France
    Nils Ternes
  199. Department of Surgical Pathology and Biopathology, Jean Perrin Comprehensive Cancer Centre, Clermont-Ferrand, France
    Nina Radosevic-Robin
  200. Johanniter GmbH - Evangelisches Krankenhaus Bethesda Mönchengladbach, West German Study Group, Mönchengladbach, Germany
    Oleg Gluz
  201. Molecular Oncology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
    Paolo Nuciforo
  202. Department of Pathology, University of Marburg, Marburg, Germany
    Paul Jank
  203. Department of Pathology and Laboratory Medicine, University of British Columbia, Columbia, USA
    Peter H. Watson
  204. Department of Anatomical Pathology, St Vincent’s Hospital Melbourne, Fitzroy, Australia
    Prudence A. Russell
  205. Cancer Immunotherapy Trials Network, Central Laboratory and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, USA
    Robert H. Pierce
  206. Clinical Trial Service Unit & Epidemiological Studies Unit, University of Oxford, Oxford, UK
    Robert Hills
  207. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
    Ruohong Shui & Wentao Yang
  208. Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
    Sabine Declercq
  209. Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Research Institute, Toronto, Canada
    Sami Tabbarah & William T. Tran
  210. Oncology Merck & Co, New Jersey, USA
    Sandra C. Souza
  211. The Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Australian Clinical Labs, Darlinghurst, Australia
    Sandra O’Toole
  212. Georgetown University Medical Center, Washington DC, USA
    Sandra Swain
  213. Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
    Scooter Willis
  214. Translational Medicine, Bristol-Myers Squibb, Princeton, USA
    Scott Ely
  215. National Surgical Adjuvant Breast and Bowel Project Operations Center/NRG Oncology, Pittsburgh, USA
    Seong- Rim Kim
  216. Anatomic Pathology, Boston, MA, USA
    Shahinaz Bedri
  217. King’s College London, London, UK
    Sheeba Irshad
  218. Guy’s Hospital, London, UK
    Sheeba Irshad
  219. Peking University First Hospital Breast Disease Center, Beijing, China
    Shi-Wei Liu
  220. Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
    Shona Hendry & Stephen B. Fox
  221. Dipartimento di Scienze della Salute (DSS), Firenze, Italy
    Simonetta Bianchi
  222. Department of Oncology, Champalimaud Clinical Centre, Lisbon, Portugal
    Sofia Bragança
  223. Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, USA
    Stephen Naber
  224. Department of Pathology, Fundación Valle del Lili, Cali, Colombia
    Sua Luz
  225. Department of Pathology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, USA
    Susan Fineberg
  226. Department of Pathology, University Hospital of Bellvitge, Oncobell, IDIBELL, L’Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
    Teresa Soler
  227. Department of Development and Regeneration, Laboratory of Experimental Urology, KU Leuven, Leuven, Belgium
    Thomas Gevaert
  228. Department of Medical Oncology, Austin Health, Heidelberg, Australia
    Tom John
  229. Department of Surgery, Kansai Medical University to Tomoharu Sugie, Breast Surgery, Kansai Medical University Hospital, Hirakata, Japan
    Tomohagu Sugie
  230. Department of Pathology, Massachusetts General Hospital, Boston, USA
    Veerle Bossuyt
  231. Pathology Department, H.U. Vall d’Hebron, Barcelona, Spain
    Vincente Peg Cámaea
  232. Division of Bioinformatics and Biostatistics, U.S. Food and Drug Administration, Wuhan, USA
    Weida Tong
  233. Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Providence, USA
    Yihong Wang
  234. Université Paris-Est, Créteil, France
    Yves Allory
  235. Praava Health, Dhaka, Bangladesh
    Zaheed Husain

Authors

  1. Mohamed Amgad
    You can also search for this author inPubMed Google Scholar
  2. Elisabeth Specht Stovgaard
    You can also search for this author inPubMed Google Scholar
  3. Eva Balslev
    You can also search for this author inPubMed Google Scholar
  4. Jeppe Thagaard
    You can also search for this author inPubMed Google Scholar
  5. Weijie Chen
    You can also search for this author inPubMed Google Scholar
  6. Sarah Dudgeon
    You can also search for this author inPubMed Google Scholar
  7. Ashish Sharma
    You can also search for this author inPubMed Google Scholar
  8. Jennifer K. Kerner
    You can also search for this author inPubMed Google Scholar
  9. Carsten Denkert
    You can also search for this author inPubMed Google Scholar
  10. Yinyin Yuan
    You can also search for this author inPubMed Google Scholar
  11. Khalid AbdulJabbar
    You can also search for this author inPubMed Google Scholar
  12. Stephan Wienert
    You can also search for this author inPubMed Google Scholar
  13. Peter Savas
    You can also search for this author inPubMed Google Scholar
  14. Leonie Voorwerk
    You can also search for this author inPubMed Google Scholar
  15. Andrew H. Beck
    You can also search for this author inPubMed Google Scholar
  16. Anant Madabhushi
    You can also search for this author inPubMed Google Scholar
  17. Johan Hartman
    You can also search for this author inPubMed Google Scholar
  18. Manu M. Sebastian
    You can also search for this author inPubMed Google Scholar
  19. Hugo M. Horlings
    You can also search for this author inPubMed Google Scholar
  20. Jan Hudeček
    You can also search for this author inPubMed Google Scholar
  21. Francesco Ciompi
    You can also search for this author inPubMed Google Scholar
  22. David A. Moore
    You can also search for this author inPubMed Google Scholar
  23. Rajendra Singh
    You can also search for this author inPubMed Google Scholar
  24. Elvire Roblin
    You can also search for this author inPubMed Google Scholar
  25. Marcelo Luiz Balancin
    You can also search for this author inPubMed Google Scholar
  26. Marie-Christine Mathieu
    You can also search for this author inPubMed Google Scholar
  27. Jochen K. Lennerz
    You can also search for this author inPubMed Google Scholar
  28. Pawan Kirtani
    You can also search for this author inPubMed Google Scholar
  29. I-Chun Chen
    You can also search for this author inPubMed Google Scholar
  30. Jeremy P. Braybrooke
    You can also search for this author inPubMed Google Scholar
  31. Giancarlo Pruneri
    You can also search for this author inPubMed Google Scholar
  32. Sandra Demaria
    You can also search for this author inPubMed Google Scholar
  33. Sylvia Adams
    You can also search for this author inPubMed Google Scholar
  34. Stuart J. Schnitt
    You can also search for this author inPubMed Google Scholar
  35. Sunil R. Lakhani
    You can also search for this author inPubMed Google Scholar
  36. Federico Rojo
    You can also search for this author inPubMed Google Scholar
  37. Laura Comerma
    You can also search for this author inPubMed Google Scholar
  38. Sunil S. Badve
    You can also search for this author inPubMed Google Scholar
  39. Mehrnoush Khojasteh
    You can also search for this author inPubMed Google Scholar
  40. W. Fraser Symmans
    You can also search for this author inPubMed Google Scholar
  41. Christos Sotiriou
    You can also search for this author inPubMed Google Scholar
  42. Paula Gonzalez-Ericsson
    You can also search for this author inPubMed Google Scholar
  43. Katherine L. Pogue-Geile
    You can also search for this author inPubMed Google Scholar
  44. Rim S. Kim
    You can also search for this author inPubMed Google Scholar
  45. David L. Rimm
    You can also search for this author inPubMed Google Scholar
  46. Giuseppe Viale
    You can also search for this author inPubMed Google Scholar
  47. Stephen M. Hewitt
    You can also search for this author inPubMed Google Scholar
  48. John M. S. Bartlett
    You can also search for this author inPubMed Google Scholar
  49. Frédérique Penault-Llorca
    You can also search for this author inPubMed Google Scholar
  50. Shom Goel
    You can also search for this author inPubMed Google Scholar
  51. Huang-Chun Lien
    You can also search for this author inPubMed Google Scholar
  52. Sibylle Loibl
    You can also search for this author inPubMed Google Scholar
  53. Zuzana Kos
    You can also search for this author inPubMed Google Scholar
  54. Sherene Loi
    You can also search for this author inPubMed Google Scholar
  55. Matthew G. Hanna
    You can also search for this author inPubMed Google Scholar
  56. Stefan Michiels
    You can also search for this author inPubMed Google Scholar
  57. Marleen Kok
    You can also search for this author inPubMed Google Scholar
  58. Torsten O. Nielsen
    You can also search for this author inPubMed Google Scholar
  59. Alexander J. Lazar
    You can also search for this author inPubMed Google Scholar
  60. Zsuzsanna Bago-Horvath
    You can also search for this author inPubMed Google Scholar
  61. Loes F. S. Kooreman
    You can also search for this author inPubMed Google Scholar
  62. Jeroen A. W. M. van der Laak
    You can also search for this author inPubMed Google Scholar
  63. Joel Saltz
    You can also search for this author inPubMed Google Scholar
  64. Brandon D. Gallas
    You can also search for this author inPubMed Google Scholar
  65. Uday Kurkure
    You can also search for this author inPubMed Google Scholar
  66. Michael Barnes
    You can also search for this author inPubMed Google Scholar
  67. Roberto Salgado
    You can also search for this author inPubMed Google Scholar
  68. Lee A. D. Cooper
    You can also search for this author inPubMed Google Scholar

Consortia

International Immuno-Oncology Biomarker Working Group

Contributions

This report is produced as a result of discussion and consensus by members of the International Immuno-Oncology Biomarker Working Group (TILs Working Group). All authors have contributed to: 1) the conception or design of the work, 2) drafting the work or revising it critically for important intellectual content, 3) final approval of the completed version, 4) accountability for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Corresponding authors

Correspondence toRoberto Salgado or Lee A. D. Cooper.

Ethics declarations

Competing interests

J.T. is funded by Visiopharm A/S, Denmark. A.M. is an equity holder in Elucid Bioimaging and in Inspirata Inc. He is also a scientific advisory consultant for Inspirata Inc. In addition he has served as a scientific advisory board member for Inspirata Inc, Astrazeneca, Bristol Meyers-Squibb and Merck. He also has sponsored research agreements with Philips and Inspirata Inc. His technology has been licensed to Elucid Bioimaging and Inspirata Inc. He is also involved in an NIH U24 grant with PathCore Inc, and three different R01 grants with Inspirata Inc. S.R.L. received travel and educational funding from Roche/Ventana. A.J.L. serves as a consultant for BMS, Merck, AZ/Medimmune, and Genentech. He is also provides consulting and advisory work for many other companies not relevant to this work. FPL does consulting for Astrazeneca, BMS, Roche, MSD Pfizer, Novartis, Sanofi, and Lilly. S.Ld.H., A.K., M.K., U.K., and M.B. are employees of Roche. J.M.S.B. is consultant for Insight Genetics, BioNTech AG, Biothernostics, Pfizer, RNA Diagnostics, and OncoXchange. He received funding from Thermo Fisher Scientific, Genoptix, Agendia, NanoString technologies, Stratifyer GmBH, and Biotheranostics. L.F.S.K. is a consultant for Roche and Novartis. J.K.K. and A.H.B. are employees of PathAI. D.L.R. is on the advisory board for Amgen, Astra Xeneca, Cell Signaling Technology, Cepheid, Daiichi Sankyo, GSK, Konica/Minolta, Merck, Nanostring, Perking Elmer, Roche/Ventana, and Ultivue. He has received research support from Astrazeneca, Cepheid, Navigate BioPharma, NextCure, Lilly, Ultivue, Roche/Ventana, Akoya/Perkin Elmer, and Nanostring. He also has financial conflicts of interest with BMS, Biocept, PixelGear, and Rarecyte. S.G. is a consultant for and/or receives funding from Eli Lilly, Novartis, and G1 Therapeutics. J.A.W.M.vdL. is a member of the scientific advisory boards of Philips, the Netherlands and ContextVision, Sweden, and receives research funding from Philips, the Netherlands and Sectra, Sweden. S.A. is a consultant for Merck, Genentech, and BMS, and receives funding from Merck, Genentech, BMS, Novartis, Celgene, and Amgen. T.O.N. has consulted for Nanostring, and has intellectual property rights and ownership interests from Bioclassifier LLC. S.L. receives research funding to her institution from Novartis, Bristol Meyers-Squibb, Merck, Roche-Genentech, Puma Biotechnology, Pfizer and Eli Lilly. She has acted as consultant (not compensated) to Seattle Genetics, Pfizer, Novartis, BMS, Merck, AstraZeneca and Roche-Genentech. She has acted as consultant (paid to her institution) to Aduro Biotech. J.H. is director and owner of Slide Score BV. M.M.S. is a medical advisory board member of OptraScan. R.S. has received research support from Merck, Roche, Puma; and travel/congress support from AstraZeneca, Roche and Merck; and he has served as an advisory board member of BMS and Roche and consults for BMS.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Cite this article

Amgad, M., Stovgaard, E.S., Balslev, E. et al. Report on computational assessment of Tumor Infiltrating Lymphocytes from the International Immuno-Oncology Biomarker Working Group.npj Breast Cancer 6, 16 (2020). https://doi.org/10.1038/s41523-020-0154-2

Download citation