1
|
|
Fanucci KA, Bai Y, Pelekanou V, Nahleh ZA, Shafi S, Burela S, Barlow WE, Sharma P, Thompson AM, Godwin AK, Rimm DL, Hortobagyi GN, Liu Y, Wang L, Wei W, Pusztai L, Blenman KRM. Image analysis-based tumor infiltrating lymphocytes measurement predicts breast cancer pathologic complete response in SWOG S0800 neoadjuvant chemotherapy trial. NPJ Breast Cancer 2023; 9:38. [PMID: 37179362 DOI: 10.1038/s41523-023-00535-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
We assessed the predictive value of an image analysis-based tumor-infiltrating lymphocytes (TILs) score for pathologic complete response (pCR) and event-free survival in breast cancer (BC). About 113 pretreatment samples were analyzed from patients with stage IIB-IIIC HER-2-negative BC randomized to neoadjuvant chemotherapy ± bevacizumab. TILs quantification was performed on full sections using QuPath open-source software with a convolutional neural network cell classifier (CNN11). We used easTILs% as a digital metric of TILs score defined as [sum of lymphocytes area (mm2)/stromal area(mm2)] × 100. Pathologist-read stromal TILs score (sTILs%) was determined following published guidelines. Mean pretreatment easTILs% was significantly higher in cases with pCR compared to residual disease (median 36.1 vs.14.8%, p < 0.001). We observed a strong positive correlation (r = 0.606, p < 0.0001) between easTILs% and sTILs%. The area under the prediction curve (AUC) was higher for easTILs% than sTILs%, 0.709 and 0.627, respectively. Image analysis-based TILs quantification is predictive of pCR in BC and had better response discrimination than pathologist-read sTILs%.
Collapse
Affiliation(s)
- Kristina A Fanucci
- Department of Internal Medicine Section of Medical Oncology and Yale Cancer Center, Yale School of Medicine, 333 Cedar St, New Haven, CT, 06520, USA
| | - Yalai Bai
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
| | - Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
- Bayer Pharmaceuticals, 245 First St Cambridge Science Center 100 and 200 Floors 1 and 2, Cambridge, MA, 02142, USA
| | - Zeina A Nahleh
- Department of Hematology/Oncology, Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL, 33331, USA
| | - Saba Shafi
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
- Department of Pathology, Ohio State University, 6100 Optometry Clinic & Health Sciences Faculty Office Building, 1664 Neil Avenue, Columbus, OH, 43210, USA
| | - Sneha Burela
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
| | - William E Barlow
- SWOG Statistics and Data Management Center, 1730 Minor Avenue Suite 1900, Seattle, WA, 98101, USA
| | - Priyanka Sharma
- Department of Medical Oncology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Alastair M Thompson
- Section of Breast Surgery, 1 Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrew K Godwin
- Department of Medical Oncology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Yihan Liu
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT, 06520, USA
| | - Leona Wang
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT, 06520, USA
| | - Wei Wei
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, CT, 06520, USA
| | - Lajos Pusztai
- Department of Internal Medicine Section of Medical Oncology and Yale Cancer Center, Yale School of Medicine, 333 Cedar St, New Haven, CT, 06520, USA
| | - Kim R M Blenman
- Department of Internal Medicine Section of Medical Oncology and Yale Cancer Center, Yale School of Medicine, 333 Cedar St, New Haven, CT, 06520, USA.
- Department of Computer Science, Yale School of Engineering and Applied Science, 17 Hillhouse Avenue, New Haven, CT, 06520, USA.
| |
Collapse
|
2
|
|
Verdicchio M, Brancato V, Cavaliere C, Isgrò F, Salvatore M, Aiello M. A pathomic approach for tumor-infiltrating lymphocytes classification on breast cancer digital pathology images. Heliyon 2023; 9:e14371. [PMID: 36950640 DOI: 10.1016/j.heliyon.2023.e14371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Background and objectives The detection of tumor-infiltrating lymphocytes (TILs) could aid in the development of objective measures of the infiltration grade and can support decision-making in breast cancer (BC). However, manual quantification of TILs in BC histopathological whole slide images (WSI) is currently based on a visual assessment, thus resulting not standardized, not reproducible, and time-consuming for pathologists. In this work, a novel pathomic approach, aimed to apply high-throughput image feature extraction techniques to analyze the microscopic patterns in WSI, is proposed. In fact, pathomic features provide additional information concerning the underlying biological processes compared to the WSI visual interpretation, thus providing more easily interpretable and explainable results than the most frequently investigated Deep Learning based methods in the literature. Methods A dataset containing 1037 regions of interest with tissue compartments and TILs annotated on 195 TNBC and HER2+ BC hematoxylin and eosin (H&E)-stained WSI was used. After segmenting nuclei within tumor-associated stroma using a watershed-based approach, 71 pathomic features were extracted from each nucleus and reduced using a Spearman's correlation filter followed by a nonparametric Wilcoxon rank-sum test and least absolute shrinkage and selection operator. The relevant features were used to classify each candidate nucleus as either TILs or non-TILs using 5 multivariable machine learning classification models trained using 5-fold cross-validation (1) without resampling, (2) with the synthetic minority over-sampling technique and (3) with downsampling. The prediction performance of the models was assessed using ROC curves. Results 21 features were selected, with most of them related to the well-known TILs properties of having regular shape, clearer margins, high peak intensity, more homogeneous enhancement and different textural pattern than other cells. The best performance was obtained by Random-Forest with ROC AUC of 0.86, regardless of resampling technique. Conclusions The presented approach holds promise for the classification of TILs in BC H&E-stained WSI and could provide support to pathologists for a reliable, rapid and interpretable clinical assessment of TILs in BC.
Collapse
Affiliation(s)
| | - Valentina Brancato
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
- Corresponding author.
| | - Carlo Cavaliere
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| | - Francesco Isgrò
- Department of Electrical Engineering and Information Technologies, University of Naples Federico II, Claudio 21, Naples, 80125, Italy
| | - Marco Salvatore
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| |
Collapse
|
3
|
|
Cserni B, Kilmartin D, O'Loughlin M, Andreu X, Bagó-Horváth Z, Bianchi S, Chmielik E, Figueiredo P, Floris G, Foschini MP, Kovács A, Heikkilä P, Kulka J, Laenkholm AV, Liepniece-Karele I, Marchiò C, Provenzano E, Regitnig P, Reiner A, Ryška A, Sapino A, Stovgaard ES, Quinn C, Zolota V, Webber M, Glynn SA, Bori R, Csörgő E, Oláh-Németh O, Pancsa T, Sejben A, Sejben I, Vörös A, Zombori T, Nyári T, Callagy G, Cserni G. ONEST (Observers Needed to Evaluate Subjective Tests) Analysis of Stromal Tumour-Infiltrating Lymphocytes (sTILs) in Breast Cancer and Its Limitations. Cancers (Basel) 2023; 15. [PMID: 36831541 DOI: 10.3390/cancers15041199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Tumour-infiltrating lymphocytes (TILs) reflect antitumour immunity. Their evaluation of histopathology specimens is influenced by several factors and is subject to issues of reproducibility. ONEST (Observers Needed to Evaluate Subjective Tests) helps in determining the number of observers that would be sufficient for the reliable estimation of inter-observer agreement of TIL categorisation. This has not been explored previously in relation to TILs. ONEST analyses, using an open-source software developed by the first author, were performed on TIL quantification in breast cancers taken from two previous studies. These were one reproducibility study involving 49 breast cancers, 23 in the first circulation and 14 pathologists in the second circulation, and one study involving 100 cases and 9 pathologists. In addition to the estimates of the number of observers required, other factors influencing the results of ONEST were examined. The analyses reveal that between six and nine observers (range 2-11) are most commonly needed to give a robust estimate of reproducibility. In addition, the number and experience of observers, the distribution of values around or away from the extremes, and outliers in the classification also influence the results. Due to the simplicity and the potentially relevant information it may give, we propose ONEST to be a part of new reproducibility analyses.
Collapse
Affiliation(s)
- Bálint Cserni
- TNG Technology Consulting GmbH, Király u. 26., 1061 Budapest, Hungary
| | - Darren Kilmartin
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Mark O’Loughlin
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Xavier Andreu
- Pathology Department, Atryshealth Co., Ltd., 08039 Barcelona, Spain
| | - Zsuzsanna Bagó-Horváth
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Simonetta Bianchi
- Division of Pathological Anatomy, Department of Health Sciences, University of Florence, 50134 Florence, Italy
| | - Ewa Chmielik
- Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Paulo Figueiredo
- Laboratório de Anatomia Patológica, IPO Coimbra, 3000-075 Coimbra, Portugal
| | - Giuseppe Floris
- Laboratory of Translational Cell & Tissue Research and KU Leuven, Department of Imaging and Pathology, Department of Pathology, University Hospitals Leuven, University of Leuven, Oude Market 13, 3000 Leuven, Belgium
| | - Maria Pia Foschini
- Unit of Anatomic Pathology, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bellaria Hospital, 40139 Bologna, Italy
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Päivi Heikkilä
- Department of Pathology, Helsinki University Central Hospital, 00029 Helsinki, Finland
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University Budapest, Üllői út 93, 1091 Budapest, Hungary
| | - Anne-Vibeke Laenkholm
- Department of Surgical Pathology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Inta Liepniece-Karele
- Department of Pathology, Riga Stradins University, Riga East Clinical University Hospital, LV-1038 Riga, Latvia
| | - Caterina Marchiò
- Unit of Pathology, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Italy
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Elena Provenzano
- Department of Histopathology, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge CB2 0QQ, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Peter Regitnig
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Angelika Reiner
- Department of Pathology, Klinikum Donaustadt, 1090 Vienna, Austria
| | - Aleš Ryška
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital, 50003 Hradec Kralove, Czech Republic
| | - Anna Sapino
- Unit of Pathology, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Italy
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | | | - Cecily Quinn
- Department of Histopathology, Irish National Breast Screening Programme, BreastCheck, St. Vincent’s University Hospital and School of Medicine, University College Dublin, D04 T6F4 Dublin, Ireland
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece
| | - Mark Webber
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Rita Bori
- Department of Pathology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
| | - Erika Csörgő
- Department of Pathology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
| | | | - Tamás Pancsa
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary
| | - Anita Sejben
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary
| | - István Sejben
- Department of Pathology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
| | - András Vörös
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary
| | - Tamás Zombori
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary
| | - Tibor Nyári
- Department of Medical Physics and Informatics, University of Szeged, 6720 Szeged, Hungary
| | - Grace Callagy
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Gábor Cserni
- Department of Pathology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
4
|
|
Miyagawa C, Nakai H, Otani T, Murakami R, Takamura S, Takaya H, Murakami K, Mandai M, Matsumura N. Histopathological subtyping of high-grade serous ovarian cancer using whole slide imaging. J Gynecol Oncol 2023. [PMID: 36807749 DOI: 10.3802/jgo.2023.34.e47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
OBJECTIVE We have established 4 histopathologic subtyping of high-grade serous ovarian cancer (HGSOC) and reported that the mesenchymal transition (MT) type has a worse prognosis than the other subtypes. In this study, we modified the histopathologic subtyping algorithm to achieve high interobserver agreement in whole slide imaging (WSI) and to characterize the tumor biology of MT type for treatment individualization. METHODS Four observers performed histopathological subtyping using WSI of HGSOC in The Cancer Genome Atlas data. As a validation set, cases from Kindai and Kyoto Universities were independently evaluated by the 4 observers to determine concordance rates. In addition, genes highly expressed in MT type were examined by gene ontology term analysis. Immunohistochemistry was also performed to validate the pathway analysis. RESULTS After algorithm modification, the kappa coefficient, which indicates interobserver agreement, was greater than 0.5 (moderate agreement) for the 4 classifications and greater than 0.7 (substantial agreement) for the 2 classifications (MT vs. non-MT). Gene expression analysis showed that gene ontology terms related to angiogenesis and immune response were enriched in the genes highly expressed in the MT type. CD31 positive microvessel density was higher in the MT type compared to the non-MT type, and tumor groups with high infiltration of CD8/CD103 positive immune cells were observed in the MT type. CONCLUSION We developed an algorithm for reproducible histopathologic subtyping classification of HGSOC using WSI. The results of this study may be useful for treatment individualization of HGSOC, including angiogenesis inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Chiho Miyagawa
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hidekatsu Nakai
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan.
| | - Tomoyuki Otani
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hisamitsu Takaya
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kosuke Murakami
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
5
|
|
Wang X, Collet L, Rediti M, Debien V, De Caluwé A, Venet D, Romano E, Rothé F, Sotiriou C, Buisseret L. Predictive Biomarkers for Response to Immunotherapy in Triple Negative Breast Cancer: Promises and Challenges. J Clin Med 2023; 12. [PMID: 36769602 DOI: 10.3390/jcm12030953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a highly heterogeneous disease with a poor prognosis and a paucity of therapeutic options. In recent years, immunotherapy has emerged as a new treatment option for patients with TNBC. However, this therapeutic evolution is paralleled by a growing need for biomarkers which allow for a better selection of patients who are most likely to benefit from this immune checkpoint inhibitor (ICI)-based regimen. These biomarkers will not only facilitate a better optimization of treatment strategies, but they will also avoid unnecessary side effects in non-responders, and limit the increasing financial toxicity linked to the use of these agents. Huge efforts have been deployed to identify predictive biomarkers for the ICI, but until now, the fruits of this labor remained largely unsatisfactory. Among clinically validated biomarkers, only programmed death-ligand 1 protein (PD-L1) expression has been prospectively assessed in TNBC trials. In addition to this, microsatellite instability and a high tumor mutational burden are approved as tumor agnostic biomarkers, but only a small percentage of TNBC fits this category. Furthermore, TNBC should no longer be approached as a single biological entity, but rather as a complex disease with different molecular, clinicopathological, and tumor microenvironment subgroups. This review provides an overview of the validated and evolving predictive biomarkers for a response to ICI in TNBC.
Collapse
|
6
|
|
Valenza C, Taurelli Salimbeni B, Santoro C, Trapani D, Antonarelli G, Curigliano G. Tumor Infiltrating Lymphocytes across Breast Cancer Subtypes: Current Issues for Biomarker Assessment. Cancers (Basel) 2023; 15. [PMID: 36765724 DOI: 10.3390/cancers15030767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) represent a surrogate biomarker of anti-tumor, lymphocyte-mediated immunity. In early, triple-negative breast cancer, TILs have level 1B of evidence to predict clinical outcomes. TILs represent a promising biomarker to select patients who can experience a better prognosis with de-intensified cancer treatments and derive larger benefits from immune checkpoint inhibitors. However, the assessment and the validation of TILs as a biomarker require a prospective and rigorous demonstration of its clinical validity and utility, provided reproducible analytical performance. With pending data about the prospective validation of TILs' clinical validity to modulate treatments in early breast cancer, this review summarizes the most important current issues and future challenges related to the implementation of TILs assessments across all breast cancer subtypes and their potential integration into clinical practice.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5748-9599
| |
Collapse
|
7
|
|
Onkar SS, Carleton NM, Lucas PC, Bruno TC, Lee AV, Vignali DAA, Oesterreich S. The Great Immune Escape: Understanding the Divergent Immune Response in Breast Cancer Subtypes. Cancer Discov 2023; 13:23-40. [PMID: 36620880 DOI: 10.1158/2159-8290.CD-22-0475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer, the most common type of cancer affecting women, encompasses a collection of histologic (mainly ductal and lobular) and molecular subtypes exhibiting diverse clinical presentation, disease trajectories, treatment options, and outcomes. Immunotherapy has revolutionized treatment for some solid tumors but has shown limited promise for breast cancers. In this review, we summarize recent advances in our understanding of the complex interactions between tumor and immune cells in subtypes of breast cancer at the cellular and microenvironmental levels. We aim to provide a perspective on opportunities for future immunotherapy agents tailored to specific features of each subtype of breast cancer. SIGNIFICANCE Although there are currently over 200 ongoing clinical trials testing immunotherapeutics, such as immune-checkpoint blockade agents, these are largely restricted to the triple-negative and HER2+ subtypes and primarily focus on T cells. With the rapid expansion of new in vitro, in vivo, and clinical data, it is critical to identify and highlight the challenges and opportunities unique for each breast cancer subtype to drive the next generation of treatments that harness the immune system.
Collapse
Affiliation(s)
- Sayali S. Onkar
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Neil M. Carleton
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter C Lucas
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA,Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Adrian V Lee
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA,Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Dario AA Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA,Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
|
Rakaee M, Adib E, Ricciuti B, Sholl LM, Shi W, Alessi JV, Cortellini A, Fulgenzi CAM, Viola P, Pinato DJ, Hashemi S, Bahce I, Houda I, Ulas EB, Radonic T, Väyrynen JP, Richardsen E, Jamaly S, Andersen S, Donnem T, Awad MM, Kwiatkowski DJ. Association of Machine Learning-Based Assessment of Tumor-Infiltrating Lymphocytes on Standard Histologic Images With Outcomes of Immunotherapy in Patients With NSCLC. JAMA Oncol 2023; 9:51-60. [PMID: 36394839 DOI: 10.1001/jamaoncol.2022.4933] [Citation(s) in RCA: 0] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Importance Currently, predictive biomarkers for response to immune checkpoint inhibitor (ICI) therapy in lung cancer are limited. Identifying such biomarkers would be useful to refine patient selection and guide precision therapy. Objective To develop a machine-learning (ML)-based tumor-infiltrating lymphocytes (TILs) scoring approach, and to evaluate TIL association with clinical outcomes in patients with advanced non-small cell lung cancer (NSCLC). Design, Setting, and Participants This multicenter retrospective discovery-validation cohort study included 685 ICI-treated patients with NSCLC with median follow-up of 38.1 and 43.3 months for the discovery (n = 446) and validation (n = 239) cohorts, respectively. Patients were treated between February 2014 and September 2021. We developed an ML automated method to count tumor, stroma, and TIL cells in whole-slide hematoxylin-eosin-stained images of NSCLC tumors. Tumor mutational burden (TMB) and programmed death ligand-1 (PD-L1) expression were assessed separately, and clinical response to ICI therapy was determined by medical record review. Data analysis was performed from June 2021 to April 2022. Exposures All patients received anti-PD-(L)1 monotherapy. Main Outcomes and Measures Objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) were determined by blinded medical record review. The area under curve (AUC) of TIL levels, TMB, and PD-L1 in predicting ICI response were calculated using ORR. Results Overall, there were 248 (56%) women in the discovery cohort and 97 (41%) in the validation cohort. In a multivariable analysis, high TIL level (≥250 cells/mm2) was independently associated with ICI response in both the discovery (PFS: HR, 0.71; P = .006; OS: HR, 0.74; P = .03) and validation (PFS: HR = 0.80; P = .01; OS: HR = 0.75; P = .001) cohorts. Survival benefit was seen in both first- and subsequent-line ICI treatments in patients with NSCLC. In the discovery cohort, the combined models of TILs/PD-L1 or TMB/PD-L1 had additional specificity in differentiating ICI responders compared with PD-L1 alone. In the PD-L1 negative (<1%) subgroup, TIL levels had superior classification accuracy for ICI response (AUC = 0.77) compared with TMB (AUC = 0.65). Conclusions and Relevance In these cohorts, TIL levels were robustly and independently associated with response to ICI treatment. Patient TIL assessment is relatively easily incorporated into the workflow of pathology laboratories at minimal additional cost, and may enhance precision therapy.
Collapse
Affiliation(s)
- Mehrdad Rakaee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Elio Adib
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weiwei Shi
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joao V Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alessio Cortellini
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Claudia A M Fulgenzi
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom.,Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - Patrizia Viola
- Department of Cellular Pathology, Imperial College London NHS Trust, London, United Kingdom
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom.,Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Sayed Hashemi
- Department of Pulmonology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Idris Bahce
- Department of Pulmonology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ilias Houda
- Department of Pulmonology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ezgi B Ulas
- Department of Pulmonology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Teodora Radonic
- Department of Pathology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Juha P Väyrynen
- Cancer and Translational Medicine Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Simin Jamaly
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Sigve Andersen
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - David J Kwiatkowski
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
9
|
|
Yi M, Wu Y, Niu M, Zhu S, Zhang J, Yan Y, Zhou P, Dai Z, Wu K. Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer. J Immunother Cancer 2022; 10. [PMID: 36460337 DOI: 10.1136/jitc-2022-005543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Agents blocking programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) have been approved for triple-negative breast cancer (TNBC). However, the response rate of anti-PD-1/PD-L1 is still unsatisfactory, partly due to immunosuppressive factors such as transforming growth factor-beta (TGF-β). In our previous pilot study, the bispecific antibody targeting TGF-β and murine PD-L1 (termed YM101) showed potent antitumor effect. In this work, we constructed a bispecific antibody targeting TGF-β and human PD-L1 (termed BiTP) and explored the antitumor effect of BiTP in TNBC. METHODS BiTP was developed using Check-BODYTM bispecific platform. The binding affinity of BiTP was measured by surface plasmon resonance, ELISA, and flow cytometry. The bioactivity was assessed by Smad and NFAT luciferase reporter assays, immunofluorescence, western blotting, and superantigen stimulation assays. The antitumor activity of BiTP was explored in humanized epithelial-mesenchymal transition-6-hPDL1 and 4T1-hPDL1 murine TNBC models. Immunohistochemical staining, flow cytometry, and bulk RNA-seq were used to investigate the effect of BiTP on immune cell infiltration. RESULTS BiTP exhibited high binding affinity to dual targets. In vitro experiments verified that BiTP effectively counteracted TGF-β-Smad and PD-L1-PD-1-NFAT signaling. In vivo animal experiments demonstrated that BiTP had superior antitumor activity relative to anti-PD-L1 and anti-TGF-β monotherapy. Mechanistically, BiTP decreased collagen deposition, enhanced CD8+ T cell penetration, and increased tumor-infiltrating lymphocytes. This improved tumor microenvironment contributed to the potent antitumor activity of BiTP. CONCLUSION BiTP retains parent antibodies' binding affinity and bioactivity, with superior antitumor activity to parent antibodies in TNBC. Our data suggest that BiTP might be a promising agent for TNBC treatment.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhang
- Wuhan YZY Biopharma Co Ltd, Wuhan, China
| | | | | | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
|
Capobianco E. Overview of triple negative breast cancer prognostic signatures in the context of data science-driven clinico-genomics research. Ann Transl Med 2022; 10:1300. [PMID: 36660729 DOI: 10.21037/atm-22-5477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
11
|
|
Zheng Q, Yang R, Ni X, Yang S, Jiao P, Wu J, Xiong L, Wang J, Jian J, Jiang Z, Wang L, Chen Z, Liu X. Quantitative Assessment of Tumor-Infiltrating Lymphocytes Using Machine Learning Predicts Survival in Muscle-Invasive Bladder Cancer. J Clin Med 2022; 11. [PMID: 36498655 DOI: 10.3390/jcm11237081] [Citation(s) in RCA: 0] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
(1) Purpose: Although assessment of tumor-infiltrating lymphocytes (TILs) has been acknowledged to have important predictive prognostic value in muscle-invasive bladder cancer (MIBC), it is limited by inter- and intra-observer variability, hampering widespread clinical application. We aimed to evaluate the prognostic value of quantitative TILs score based on a machine learning (ML) algorithm to identify MIBC patients who might benefit from immunotherapy or the de-escalation of therapy. (2) Methods: We constructed an artificial neural network classifier for tumor cells, lymphocytes, stromal cells, and “ignore” cells from hematoxylin-and-eosin-stained slide images using the QuPath open source software. We defined four unique TILs variables based on ML to analyze TILs measurements. Pathological slide images from 133 MIBC patients were retrospectively collected as the discovery set to determine the optimal association of ML-read TILs variables with patient survival outcomes. For validation, we evaluated an independent external validation set consisting of 247 MIBC patients. (3) Results: We found that all four TILs variables had significant prognostic associations with survival outcomes in MIBC patients (p < 0.001 for all comparisons), with higher TILs score being associated with better prognosis. Univariate and multivariate Cox regression analyses demonstrated that electronic TILs (eTILs) variables were independently associated with overall survival after adjustment for clinicopathological factors including age, sex, and pathological stage (p < 0.001 for all analyses). Results analyzed in different subgroups showed that the eTILs variable was a strong prognostic factor that was not redundant with pre-existing clinicopathological features (p < 0.05 for all analyses). (4) Conclusion: ML-driven cell classifier-defined TILs variables were robust and independent prognostic factors in two independent cohorts of MIBC patients. eTILs have the potential to identify a subset of high-risk stage II or stage III-IV MIBC patients who might benefit from adjuvant immunotherapy.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Song Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiejun Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lin Xiong
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhengyu Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (Z.C.); (X.L.)
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (Z.C.); (X.L.)
| |
Collapse
|
12
|
|
Cho HG, Cho SI, Choi S, Jung W, Shin J, Park G, Moon J, Ma M, Song H, Mostafavi M, Kang M, Pereira S, Paeng K, Yoo D, Ock CY, Kim S. Artificial Intelligence-Powered Whole-Slide Image Analyzer Reveals a Distinctive Distribution of Tumor-Infiltrating Lymphocytes in Neuroendocrine Neoplasms. Diagnostics (Basel) 2022; 12. [PMID: 36292028 DOI: 10.3390/diagnostics12102340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Despite the importance of tumor-infiltrating lymphocytes (TIL) and PD-L1 expression to the immune checkpoint inhibitor (ICI) response, a comprehensive assessment of these biomarkers has not yet been conducted in neuroendocrine neoplasm (NEN). We collected 218 NENs from multiple organs, including 190 low/intermediate-grade NENs and 28 high-grade NENs. TIL distribution was derived from Lunit SCOPE IO, an artificial intelligence (AI)-powered hematoxylin and eosin (H&E) analyzer, as developed from 17,849 whole slide images. The proportion of intra-tumoral TIL-high cases was significantly higher in high-grade NEN (75.0% vs. 46.3%, p = 0.008). The proportion of PD-L1 combined positive score (CPS) ≥ 1 case was higher in high-grade NEN (85.7% vs. 33.2%, p < 0.001). The PD-L1 CPS ≥ 1 group showed higher intra-tumoral, stromal, and combined TIL densities, compared to the CPS < 1 group (7.13 vs. 2.95, p < 0.001; 200.9 vs. 120.5, p < 0.001; 86.7 vs. 56.1, p = 0.004). A significant correlation was observed between TIL density and PD-L1 CPS (r = 0.37, p < 0.001 for intra-tumoral TIL; r = 0.24, p = 0.002 for stromal TIL and combined TIL). AI-powered TIL analysis reveals that intra-tumoral TIL density is significantly higher in high-grade NEN, and PD-L1 CPS has a positive correlation with TIL densities, thus showing its value as predictive biomarkers for ICI response in NEN.
Collapse
|
13
|
|
Kolberg-Liedtke C, Feuerhake F, Garke M, Christgen M, Kates R, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Kuemmel S, Wuerstlein R, Graeser M, Nitz U, Kreipe H, Gluz O, Harbeck N. Impact of stromal tumor-infiltrating lymphocytes (sTILs) on response to neoadjuvant chemotherapy in triple-negative early breast cancer in the WSG-ADAPT TN trial. Breast Cancer Res 2022; 24:58. [PMID: 36056374 DOI: 10.1186/s13058-022-01552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Higher density of stromal tumor-infiltrating lymphocytes (sTILs) at baseline has been associated with increased rates of pathological complete response (pCR) after neoadjuvant chemotherapy (NACT) in triple-negative breast cancer (TNBC). While evidence supports favorable association of pCR with survival in TNBC, an independent impact of sTILs (after adjustment for pCR) on survival is not yet established. Moreover, the impact of sTIL dynamics during NACT on pCR and survival in TNBC is unknown. METHODS The randomized WSG-ADAPT TN phase II trial compared efficacy of 12-week nab-paclitaxel with gemcitabine versus carboplatin. This preplanned translational analysis assessed impacts of sTIL measurements at baseline (sTIL-0) and after 3 weeks of chemotherapy (sTIL-3) on pCR and invasive disease-free survival (iDFS). Predictive performance of sTIL-0 and sTIL-3 for pCR was quantified by ROC analysis and logistic regression; Kaplan-Meier estimation and Cox regression (with mediation analysis) were used to determine their impact on iDFS. RESULTS For prediction of pCR, the AUC statistics for sTIL-0 and sTIL-3 were 0.60 and 0.63, respectively, in all patients; AUC for sTIL-3 was higher in NP/G. The positive predictive value (PPV) of "lymphocyte-predominant" status (sTIL-0 ≥ 60%) at baseline was 59.3%, though only 13.0% of patients had this status. To predict non-pCR, the cut point sTIL-0 ≤ 10% yielded PPV = 69.5% while addressing 33.8% of patients. Higher sTIL levels (particularly at 3 weeks) were independently and favorably associated with better iDFS, even after adjusting for pCR. For example, the adjusted hazard ratio for 3-week sTILs ≥ 60% (vs. < 60%) was 0.48 [0.23-0.99]. Low cellularity in 3-week biopsies was the strongest individual predictor for pCR (in both therapy arms), but not for iDFS. CONCLUSION The independent impact of sTILs on iDFS suggests that favorable immune response can influence key tumor biological processes for long-term survival. The results suggest that the reliability of pCR following neoadjuvant therapy as a surrogate for survival could vary among subgroups in TNBC defined by immune response or other factors. Dynamic measurements of sTILs under NACT could support immune response-guided patient selection for individualized therapy approaches for both very low levels (more effective therapies) and very high levels (de-escalation concepts). TRIAL REGISTRATION Clinical trials No: NCT01815242, retrospectively registered January 25, 2013.
Collapse
Affiliation(s)
- Cornelia Kolberg-Liedtke
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | | | | | | | - Ronald Kates
- West German Study Group, Mönchengladbach, Germany
| | | | | | - Michael Braun
- Breast Center, Rotkreuz Clinics Munich, Munich, Germany
| | - Mathias Warm
- Breast Center, City Hospital Holweide, Cologne, Germany
| | | | | | - Bahriye Aktas
- Department of Gynecology, University Hospital Leipzig, Leipzig, Germany
| | | | - Sherko Kuemmel
- West German Study Group, Mönchengladbach, Germany.,Breast Unit, Kliniken Essen-Mitte, Essen, Germany.,Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rachel Wuerstlein
- West German Study Group, Mönchengladbach, Germany.,Breast Center, LMU University Hospital, Munich, Germany
| | - Monika Graeser
- West German Study Group, Mönchengladbach, Germany.,University Hospital Hamburg-Eppendorf, Hamburg, Germany.,Breast Center Niederrhein, Ev. Hospital Bethesda, Mönchengladbach, Germany
| | - Ulrike Nitz
- West German Study Group, Mönchengladbach, Germany.,Breast Center Niederrhein, Ev. Hospital Bethesda, Mönchengladbach, Germany
| | - Hans Kreipe
- Institute of Pathology, Medical School Hannover, Hannover, Germany
| | - Oleg Gluz
- West German Study Group, Mönchengladbach, Germany.,Breast Center Niederrhein, Ev. Hospital Bethesda, Mönchengladbach, Germany
| | - Nadia Harbeck
- West German Study Group, Mönchengladbach, Germany.,Breast Center, LMU University Hospital, Munich, Germany
| |
Collapse
|
14
|
|
Su T, Wang S, Huang S, Cai H, McKinley ET, Beeghly-Fadiel A, Zheng W, Shu XO, Cai Q. Multiplex immunohistochemistry and high-throughput image analysis for evaluation of spatial tumor immune cell markers in human breast cancer. Cancer Biomark 2022. [PMID: 36093688 DOI: 10.3233/CBM-220071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The clinicopathological significance of spatial tumor-infiltrating lymphocytes (TILs) subpopulations is not well studied due to lack of high-throughput scalable methodology for studies with large human sample sizes. OBJECTIVE Establishing a cyclic fluorescent multiplex immunohistochemistry (mIHC/IF) method coupled with computer-assisted high-throughput quantitative analysis to evaluate associations of six TIL markers (CD3, CD8, CD20, CD56, FOXP3, and PD-L1) with clinicopathological factors of breast cancer. METHODS Our 5-plex mIHC/IF staining was shown to be reliable and highly sensitive for labeling three biomarkers per tissue section. Through repetitive cycles of 5-plex mIHC/IF staining, more than 12 biomarkers could be detected per single tissue section. Using open-source software CellProfiler, the measurement pipelines were successfully developed for high-throughput multiplex evaluation of intratumoral and stromal TILs. RESULTS In analyses of 188 breast cancer samples from the Nashville Breast Health Study, high-grade tumors showed significantly increased intratumoral CD3+CD8+ CTL density (P= 0.0008, false discovery rate (FDR) adjusted P= 0.0168) and intratumoral PD-L1 expression (P= 0.0061, FDR adjusted P= 0.0602) compared with low-grade tumors. CONCLUSIONS The high- and low-grade breast cancers exhibit differential immune responses which may have clinical significances. The multiplexed imaging quantification strategies established in this study are reliable, cost-efficient and applicable in regular laboratory settings for high-throughput tissue biomarker studies, especially retrospective and population-based studies using archived paraffin tissues.
Collapse
Affiliation(s)
- Timothy Su
- Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA.,Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Shuyang Wang
- Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA.,Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai, China.,Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Shuya Huang
- Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA.,Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Cai
- Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Eliot T McKinley
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Cell and Development Biology, Vanderbilt University, Nashville, TN, USA
| | - Alicia Beeghly-Fadiel
- Department of Medicine, Division of Epidemiology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | |
|