1
|
Monette A, Warren S, Barrett JC, Garnett-Benson C, Schalper KA, Taube JM, Topp B, Snyder A. Biomarker development for PD-(L)1 axis inhibition: a consensus view from the SITC Biomarkers Committee. J Immunother Cancer 2024; 12:e009427. [PMID: 39032943 PMCID: PMC11261685 DOI: 10.1136/jitc-2024-009427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Therapies targeting the programmed cell death protein-1/programmed death-ligand 1 (PD-L1) (abbreviated as PD-(L)1) axis are a significant advancement in the treatment of many tumor types. However, many patients receiving these agents fail to respond or have an initial response followed by cancer progression. For these patients, while subsequent immunotherapies that either target a different axis of immune biology or non-immune combination therapies are reasonable treatment options, the lack of predictive biomarkers to follow-on agents is impeding progress in the field. This review summarizes the current knowledge of mechanisms driving resistance to PD-(L)1 therapies, the state of biomarker development along this axis, and inherent challenges in future biomarker development for these immunotherapies. Innovation in the development and application of novel biomarkers and patient selection strategies for PD-(L)1 agents is required to accelerate the delivery of effective treatments to the patients most likely to respond.
Collapse
Affiliation(s)
- Anne Monette
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
| | | | | | | | | | - Janis M Taube
- The Mark Foundation Center for Advanced Genomics and Imaging at Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
2
|
Bachmann C. New Achievements from Molecular Biology and Treatment Options for Refractory/Relapsed Ovarian Cancer-A Systematic Review. Cancers (Basel) 2023; 15:5356. [PMID: 38001616 PMCID: PMC10669965 DOI: 10.3390/cancers15225356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Ovarian cancer (OC) has a high rate of mortality and is the fifth most common cause of death in females all over the world. The etiology is still unclear. Numerous factors such as smoking, obesity, and unhealthy diet may affect the risk of OC. Having a family history of breast and OC is one of the main risks for developing OC. Mutations of BRCA1/2 are associated with OC risk as well. The histopathological classification of OC reveals the four most common types: serous, clear cell, endometrioid, and mucinous; these are epithelial OC types, and other types are rare. Furthermore, OC can be subdivided into types I and II. Type I tumors are most probably caused by atypical proliferative tumors. Type II tumors include high-grade carcinoma of the serous type, carcinosarcoma, and carcinoma, which are not differentiated and generally originate from tubal intraepithelial carcinoma of the serous type. Typically, type I tumors are present in early stages, usually with good prognosis. Type II tumors are classified as high-grade tumors and are most often diagnosed at advanced FIGO stages with poor prognosis. High-grade serous OC accounts for 90% of serous OC. Tumor heterogeneity aggravates OC treatment. The standard care for primary epithelial ovarian cancer (EOC) is cytoreductive surgery followed by platinum-based chemotherapy. Neoadjuvant chemotherapy can be used in certain cases followed by cytoreductive surgery. The main prognostic factor is complete tumor resection. However, about 70% of patients relapse. Resistance to chemotherapeutic agents remains a major challenge in EOC treatment, in which many different factors are involved. In recent years, the examination of molecular parameters and their prognostic impact has become increasingly relevant in EOC, and furthermore, the use of immunotherapy has expanded the therapeutic range. As the clinical need is greatest for relapsed patients, this systematic review will focus on recent advances in molecular biology with prognostic and predictive markers and treatment options for recurrent/refractory OC. Inclusion criteria for the review: potential prospective or predictive biomarkers in preclinical or clinical use in relapsed and refractory OC, prognostic impact, clinical and preclinical trials, and immunotherapy. Exclusion criteria for the review: primary OC, no full text or abstract available, not the topic mentioned above, and text not available in English. Risk of bias: the included studies were evaluated descriptively for the topics mentioned above, and data were not compared with each other. The objective is to highlight the molecular mechanisms of the most promising targeted agents under clinical investigation to demonstrate their potential relevance in recurrent/refractory OC.
Collapse
Affiliation(s)
- Cornelia Bachmann
- Department of Womens' Health, University Tübingen, 72070 Tübingen, Germany
| |
Collapse
|
3
|
Barrera C, Corredor G, Viswanathan VS, Ding R, Toro P, Fu P, Buzzy C, Lu C, Velu P, Zens P, Berezowska S, Belete M, Balli D, Chang H, Baxi V, Syrigos K, Rimm DL, Velcheti V, Schalper K, Romero E, Madabhushi A. Deep computational image analysis of immune cell niches reveals treatment-specific outcome associations in lung cancer. NPJ Precis Oncol 2023; 7:52. [PMID: 37264091 PMCID: PMC10235089 DOI: 10.1038/s41698-023-00403-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/19/2023] [Indexed: 06/03/2023] Open
Abstract
The tumor immune composition influences prognosis and treatment sensitivity in lung cancer. The presence of effective adaptive immune responses is associated with increased clinical benefit after immune checkpoint blockers. Conversely, immunotherapy resistance can occur as a consequence of local T-cell exhaustion/dysfunction and upregulation of immunosuppressive signals and regulatory cells. Consequently, merely measuring the amount of tumor-infiltrating lymphocytes (TILs) may not accurately reflect the complexity of tumor-immune interactions and T-cell functional states and may not be valuable as a treatment-specific biomarker. In this work, we investigate an immune-related biomarker (PhenoTIL) and its value in associating with treatment-specific outcomes in non-small cell lung cancer (NSCLC). PhenoTIL is a novel computational pathology approach that uses machine learning to capture spatial interplay and infer functional features of immune cell niches associated with tumor rejection and patient outcomes. PhenoTIL's advantage is the computational characterization of the tumor immune microenvironment extracted from H&E-stained preparations. Association with clinical outcome and major non-small cell lung cancer (NSCLC) histology variants was studied in baseline tumor specimens from 1,774 lung cancer patients treated with immunotherapy and/or chemotherapy, including the clinical trial Checkmate 057 (NCT01673867).
Collapse
Affiliation(s)
- Cristian Barrera
- Department of Biomedical Engineering, School of Medicine, Emory University, Atlanta, GA, USA
| | - Germán Corredor
- Department of Biomedical Engineering, School of Medicine, Emory University, Atlanta, GA, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Ruiwen Ding
- Case Western Reserve University, School of Engineering, Cleveland, OH, USA
| | | | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Christina Buzzy
- Case Western Reserve University, School of Engineering, Cleveland, OH, USA
| | - Cheng Lu
- Department of Biomedical Engineering, School of Medicine, Emory University, Atlanta, GA, USA
| | - Priya Velu
- Weill Cornell Medical College, New York, NY, USA
| | - Philipp Zens
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Sabina Berezowska
- Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | | | - Han Chang
- Bristol Myers Squibb, New York, NY, USA
| | | | - Konstantinos Syrigos
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - David L Rimm
- School of Medicine, Yale University, New Haven, CT, USA
| | | | - Kurt Schalper
- School of Medicine, Yale University, New Haven, CT, USA
| | - Eduardo Romero
- Universidad Nacional de Colombia, Facultad de Medicina, Bogotá, Colombia
| | - Anant Madabhushi
- Department of Biomedical Engineering, School of Medicine, Emory University, Atlanta, GA, USA.
- VA Medical Center, Atlanta, OH, USA.
| |
Collapse
|
4
|
Kwok HH, Yang J, Lam DCL. Breaking the Invisible Barriers: Unleashing the Full Potential of Immune Checkpoint Inhibitors in Oncogene-Driven Lung Adenocarcinoma. Cancers (Basel) 2023; 15:2749. [PMID: 37345086 DOI: 10.3390/cancers15102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The rapid development of targeted therapy paved the way toward personalized medicine for advanced non-small cell lung cancer (NSCLC). Lung adenocarcinoma (ADC) harboring actionable genetic alternations including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), Kirsten rat sarcoma virus (ALK) and c-ros oncogene 1 (ROS1) treated with tyrosine kinase inhibitors (TKIs) incurred lesser treatment toxicity but better therapeutic responses compared with systemic chemotherapy. Angiogenesis inhibitors targeting vascular endothelial growth factor (VEGF) have also shown an increase in overall survival (OS) for NSCLC patients. However, acquired resistance to these targeted therapies remains a major obstacle to long-term maintenance treatment for lung ADC patients. The emergence of immune checkpoint inhibitors (ICIs) against programmed cell death protein 1 (PD-1) or programmed cell death-ligand 1 (PD-L1) has changed the treatment paradigm for NSCLC tumors without actionable genetic alternations. Clinical studies have suggested, however, that there are no survival benefits with the combination of targeted therapy and ICIs. In this review, we will summarize and discuss the current knowledge on the tumor immune microenvironment and the dynamics of immune phenotypes, which could be crucial in extending the applicability of ICIs for this subpopulation of lung ADC patients.
Collapse
Affiliation(s)
- Hoi-Hin Kwok
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jiashuang Yang
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Chi-Leung Lam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Wu CG, Casanova R, Mairinger F, Soltermann A. Lung adenocarcinoma patients with malignant pleural effusions in hot adaptive immunity status have a longer overall survival. Front Oncol 2022; 12:1031094. [PMID: 36267973 PMCID: PMC9577289 DOI: 10.3389/fonc.2022.1031094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant pleural effusion (MPE) is a common complication of lung adenocarcinoma (LADC) which is associated with a dismal prognosis. We investigated the prognostic role of PD-L1 and other immunomodulators expression in the immune compartment of MPE immune composition. MPE cytologic cell blocks of 83 LADC patients were analysed for the mRNA expression of 770 cancer-immune genes by the NanoString nCounter platform. The expression of relevant immune cell lineage markers was validated by immunohistochemistry (IHC) using quantitative pathology. The mRNA immune profiling identified four MPE patient clusters (C). C1/2 (adaptive+, hot) showed better overall survival (OS) than C3/4 (adaptive-, cold). Additionally, cold immunity profiles (adaptive-), C4 (innate+) were associated with worse OS than C3 (innate-). High PD-L1 expression was linked to the regulation of T cell activation and interferon signalling pathways. Genes of pattern recognition receptor and type I interferon signalling pathways were specifically upregulated in the long-survival (≥90 days) patient group. Moreover, immunomodulators were co-activated and highly expressed in hot adaptive immunity patient clusters, whereas CD274 (PD-L1), TNFRSF9 (4-1BB), VEGFA (VEGF-A) and CD276 (B7-H3) were upregulated in the groups referred as cold. The patient cluster, age and PD-L1 expression were independent prognosticators for LADC MPE patients (p-value < 0.05). Our study sheds light on the variances of immune contexture regarding different PD-L1 expression and survival conditions. It revealed four distinct prognostic patient clusters with specific immune cell components and immunomodulator expression profiles, which, collectively, is supportive for future therapeutic and prognosis for cancer management.
Collapse
Affiliation(s)
- Cheng-Guang Wu
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| | - Ruben Casanova
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Alex Soltermann
- Facharzt Foederatio Medicorum Helveticorum (FMH) Pathologie, Pathologie Länggasse, Ittigen, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| |
Collapse
|
6
|
Freudenstein D, Litchfield C, Caramia F, Wright G, Solomon BJ, Ball D, Keam SP, Neeson P, Haupt Y, Haupt S. TP53 Status, Patient Sex, and the Immune Response as Determinants of Lung Cancer Patient Survival. Cancers (Basel) 2020; 12:cancers12061535. [PMID: 32545367 PMCID: PMC7352604 DOI: 10.3390/cancers12061535] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
Lung cancer poses the greatest cancer-related death risk and males have poorer outcomes than females, for unknown reasons. Patient sex is not a biological variable considered in lung cancer standard of care. Correlating patient genetics with outcomes is predicted to open avenues for improved management. Using a bioinformatics approach across non-small cell lung cancer (NSCLC) subtypes, we identified where patient sex, mutation of the major tumor suppressor gene, Tumour protein P53 (TP53), and immune signatures stratified outcomes in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), among datasets of The Cancer Genome Atlas (TCGA). We exposed sex and TP53 gene mutations as prognostic for LUAD survival. Longest survival in LUAD occurred among females with wild-type (wt) TP53 genes, high levels of immune infiltration and enrichment for pathway signatures of Interferon Gamma (INF-γ), Tumour Necrosis Factor (TNF) and macrophages-monocytes. In contrast, poor survival in men with LUAD and wt TP53 genes corresponded with enrichment of Transforming Growth Factor Beta 1 (TGFB1, hereafter TGF-β) and wound healing signatures. In LUAD with wt TP53 genes, elevated gene expression of immune checkpoint CD274 (hereafter: PD-L1) and also protein 53 (p53) negative-regulators of the Mouse Double Minute (MDM)-family predict novel avenues for combined immunotherapies. LUSC is dominated by male smokers with TP53 gene mutations, while a minor population of TCGA LC patients with wt TP53 genes unexpectedly had the poorest survival, suggestive of a separate etiology. We conclude that advanced approaches to LUAD and LUSC therapy lie in the consideration of patient sex, TP53 gene mutation status and immune signatures.
Collapse
Affiliation(s)
- Donald Freudenstein
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
| | - Cassandra Litchfield
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
| | - Franco Caramia
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
| | - Gavin Wright
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia;
| | - Benjamin J. Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
| | - David Ball
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
- Department of Radiation Oncology Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Simon P. Keam
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
- Cancer Immunology Research, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Paul Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
- Cancer Immunology Research, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Ygal Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3004, Australia
| | - Sue Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia; (D.F.); (C.L.); (F.C.); (S.P.K.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; (D.B.); (P.N.)
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence:
| |
Collapse
|
7
|
Neagu M, Bostan M, Constantin C. Protein microarray technology: Assisting personalized medicine in oncology (Review). WORLD ACADEMY OF SCIENCES JOURNAL 2019. [DOI: 10.3892/wasj.2019.15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Monica Neagu
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| | - Marinela Bostan
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| | - Carolina Constantin
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
8
|
Abstract
Two graph theoretic concepts—clique and bipartite graphs—are explored to identify the network biomarkers for cancer at the gene network level. The rationale is that a group of genes work together by forming a cluster or a clique-like structures to initiate a cancer. After initiation, the disease signal goes to the next group of genes related to the second stage of a cancer, which can be represented as a bipartite graph. In other words, bipartite graphs represent the cross-talk among the genes between two disease stages. To prove this hypothesis, gene expression values for three cancers— breast invasive carcinoma (BRCA), colorectal adenocarcinoma (COAD) and glioblastoma multiforme (GBM)—are used for analysis. First, a co-expression gene network is generated with highly correlated gene pairs with a Pearson correlation coefficient ≥ 0.9. Second, clique structures of all sizes are isolated from the co-expression network. Then combining these cliques, three different biomarker modules are developed—maximal clique-like modules, 2-clique-1-bipartite modules, and 3-clique-2-bipartite modules. The list of biomarker genes discovered from these network modules are validated as the essential genes for causing a cancer in terms of network properties and survival analysis. This list of biomarker genes will help biologists to design wet lab experiments for further elucidating the complex mechanism of cancer.
Collapse
|
9
|
Monette A, Morou A, Al-Banna NA, Rousseau L, Lattouf JB, Rahmati S, Tokar T, Routy JP, Cailhier JF, Kaufmann DE, Jurisica I, Lapointe R. Failed immune responses across multiple pathologies share pan-tumor and circulating lymphocytic targets. J Clin Invest 2019; 129:2463-2479. [PMID: 30912767 DOI: 10.1172/jci125301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rationale Tumor infiltrating lymphocytes are widely associated with positive outcomes, yet carry key indicators of a systemic failed immune response against unresolved cancer. Cancer immunotherapies can reverse their tolerance phenotypes, while preserving tumor-reactivity and neoantigen-specificity shared with circulating immune cells. Objectives We performed comprehensive transcriptomic analyses to identify gene signatures common to circulating and tumor infiltrating lymphocytes in the context of clear cell renal cell carcinoma. Modulated genes also associated with disease outcome were validated in other cancer types. Findings Using bioinformatics, we identified practical diagnostic markers and actionable targets of the failed immune response. On circulating lymphocytes, three genes, LEF1, FASLG, and MMP9, could efficiently stratify patients from healthy control donors. From their associations with resistance to cancer immunotherapies and microbial infections, we uncovered not only pan-cancer, but pan-pathology failed immune response profiles. A prominent lymphocytic matrix metallopeptidase cell migration pathway, is central to a panoply of diseases and tumor immunogenicity, correlates with multi-cancer recurrence, and identifies a feasible, non-invasive approach to pan-pathology diagnoses. Conclusions The non-invasive differently expressed genes we have identified warrant future investigation towards the development of their potential in precision diagnostics and precision pan-disease immunotherapeutics.
Collapse
Affiliation(s)
- Anne Monette
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Antigoni Morou
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Nadia A Al-Banna
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Basic Medical Sciences, College of Medicine, QU Health Cluster, Qatar University, Doha, Qatar
| | - Louise Rousseau
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada
| | - Jean-Baptiste Lattouf
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Sara Rahmati
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Tomas Tokar
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jean-François Cailhier
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Nephrology Division, Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Daniel E Kaufmann
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Igor Jurisica
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada.,Department of Medical Biophysics and.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Slovak Republic
| | - Réjean Lapointe
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|