1
|
Federico A, Pavel A, Möbus L, McKean D, Del Giudice G, Fortino V, Niehues H, Rastrick J, Eyerich K, Eyerich S, van den Bogaard E, Smith C, Weidinger S, de Rinaldis E, Greco D. The integration of large-scale public data and network analysis uncovers molecular characteristics of psoriasis. Hum Genomics 2022; 16:62. [PMID: 36437479 PMCID: PMC9703794 DOI: 10.1186/s40246-022-00431-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/07/2022] [Indexed: 11/29/2022] Open
Abstract
In recent years, a growing interest in the characterization of the molecular basis of psoriasis has been observed. However, despite the availability of a large amount of molecular data, many pathogenic mechanisms of psoriasis are still poorly understood. In this study, we performed an integrated analysis of 23 public transcriptomic datasets encompassing both lesional and uninvolved skin samples from psoriasis patients. We defined comprehensive gene co-expression network models of psoriatic lesions and uninvolved skin. Moreover, we curated and exploited a wide range of functional information from multiple public sources in order to systematically annotate the inferred networks. The integrated analysis of transcriptomics data and co-expression networks highlighted genes that are frequently dysregulated and show aberrant patterns of connectivity in the psoriatic lesion compared with the unaffected skin. Our approach allowed us to also identify plausible, previously unknown, actors in the expression of the psoriasis phenotype. Finally, we characterized communities of co-expressed genes associated with relevant molecular functions and expression signatures of specific immune cell types associated with the psoriasis lesion. Overall, integrating experimental driven results with curated functional information from public repositories represents an efficient approach to empower knowledge generation about psoriasis and may be applicable to other complex diseases.
Collapse
Affiliation(s)
- Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- BioMeditech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
- Tampere Institute for Advanced Studies, Tampere University, Tampere, Finland
| | - Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- BioMeditech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- BioMeditech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - David McKean
- Sanofi Immunology and Inflammation Research Therapeutic Area, Precision Immunology Cluster, Cambridge, Massachusetts, USA
| | - Giusy Del Giudice
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- BioMeditech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Hanna Niehues
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Joe Rastrick
- Immunology Therapeutic Area, UCB Pharma, Slough, UK
| | - Kilian Eyerich
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Stefanie Eyerich
- ZAUM-Center of Allergy and Environment, Technical University and Helmholtz Center Munich, Munich, Germany
| | - Ellen van den Bogaard
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Catherine Smith
- St. John's Institute of Dermatology, King's College London, London, UK
| | | | - Emanuele de Rinaldis
- Sanofi Immunology and Inflammation Research Therapeutic Area, Precision Immunology Cluster, Cambridge, Massachusetts, USA
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön Katu 34, 33520, Tampere, Finland.
- BioMeditech Institute, Tampere University, Tampere, Finland.
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland.
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Ledsgaard L, Ljungars A, Rimbault C, Sørensen CV, Tulika T, Wade J, Wouters Y, McCafferty J, Laustsen AH. Advances in antibody phage display technology. Drug Discov Today 2022; 27:2151-2169. [PMID: 35550436 DOI: 10.1016/j.drudis.2022.05.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 01/06/2023]
Abstract
Phage display technology can be used for the discovery of antibodies for research, diagnostic, and therapeutic purposes. In this review, we present and discuss key parameters that can be optimized when performing phage display selection campaigns, including the use of different antibody formats and advanced strategies for antigen presentation, such as immobilization, liposomes, nanodiscs, virus-like particles, and whole cells. Furthermore, we provide insights into selection strategies that can be used for the discovery of antibodies with complex binding requirements, such as targeting a specific epitope, cross-reactivity, or pH-dependent binding. Lastly, we provide a description of specialized phage display libraries for the discovery of bispecific antibodies and pH-sensitive antibodies. Together, these methods can be used to improve antibody discovery campaigns against all types of antigen. Teaser: This review provides an overview of the different strategies that can be exploited to improve the success rate of antibody phage display discovery campaigns, addressing key parameters, such as antigen presentation, selection methodologies, and specialized libraries.
Collapse
Affiliation(s)
- Line Ledsgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark.
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Charlotte Rimbault
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Tulika Tulika
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Jack Wade
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Yessica Wouters
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - John McCafferty
- Department of Medicine, Addenbrookes Hospital, Box 157, Hills Road, Cambridge, CB2 0QQ, UK; Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
3
|
Antibody Identification for Antigen Detection in Formalin-Fixed Paraffin-Embedded Tissue Using Phage Display and Naïve Libraries. Antibodies (Basel) 2021; 10:antib10010004. [PMID: 33466676 PMCID: PMC7839037 DOI: 10.3390/antib10010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Immunohistochemistry is a widely used technique for research and diagnostic purposes that relies on the recognition by antibodies of antigens expressed in tissues. However, tissue processing and particularly formalin fixation affect the conformation of these antigens through the formation of methylene bridges. Although antigen retrieval techniques can partially restore antigen immunoreactivity, it is difficult to identify antibodies that can recognize their target especially in formalin-fixed paraffin-embedded tissues. Most of the antibodies currently used in immunohistochemistry have been obtained by animal immunization; however, in vitro display techniques represent alternative strategies that have not been fully explored yet. This review provides an overview of phage display-based antibody selections using naïve antibody libraries on various supports (fixed cells, dissociated tissues, tissue fragments, and tissue sections) that have led to the identification of antibodies suitable for immunohistochemistry.
Collapse
|
4
|
van den Akker GGH, Eijssen LMT, Richardson SM, Rhijn LWV, Hoyland JA, Welting TJM, Voncken JW. A Membranome-Centered Approach Defines Novel Biomarkers for Cellular Subtypes in the Intervertebral Disc. Cartilage 2020; 11:203-220. [PMID: 29629573 PMCID: PMC7097986 DOI: 10.1177/1947603518764260] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Lack of specific marker-sets prohibits definition and functional distinction of cellular subtypes in the intervertebral disc (IVD), such as those from the annulus fibrosus (AF) and the nucleus pulposus (NP). DESIGN We recently generated immortalized cell lines from human NP and AF tissues; these comprise a set of functionally distinct clonal subtypes. Whole transcriptome analyses were performed of 12 phenotypically distinct clonal cell lines (4× NP-Responder, 4× NP-nonResponder, 2× AF-Sheet forming, and 2× AF-nonSheet forming). Data sets were filtered for membrane-associated marker genes and compared to literature. RESULTS Comparison of our immortal cell lines to published primary NP, AF, and articular chondrocytes (AC) transcriptome datasets revealed preservation of AF and NP phenotypes. NP-specific membrane-associated genes were defined by comparison to AF cells in both the primary dataset (46 genes) and immortal cell-lines (161 genes). Definition of AF-specific membrane-associated genes yielded 125 primary AF cell and 92 immortal cell-line markers. Overlap between primary and immortal NP cells yielded high-confidence NP-specific marker genes for NP-R (CLDN11, TMEFF2, CA12, ANXA2, CD44) and NP-nR (EFNA1, NETO2, SLC2A1). Overlap between AF and immortal AF subtypes yielded specific markers for AF-S (COLEC12, LPAR1) and AF-nS (CHIC1). CONCLUSIONS The current study provides a reference platform for preclinical evaluation of novel membrane-associated cell type-specific markers in the IVD. Future research will focus on their biological relevance for IVD function in development, homeostasis, and degenerate conditions.
Collapse
Affiliation(s)
- Guus G. H. van den Akker
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Lars M. T. Eijssen
- Department of Bioinformatics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Stephen M. Richardson
- Centre for Regenerative Medicine, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Lodewijk W. van Rhijn
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Judith A. Hoyland
- Centre for Regenerative Medicine, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Tim J. M. Welting
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jan Willem Voncken
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
5
|
Patel N, Weekes D, Drosopoulos K, Gazinska P, Noel E, Rashid M, Mirza H, Quist J, Brasó-Maristany F, Mathew S, Ferro R, Pereira AM, Prince C, Noor F, Francesch-Domenech E, Marlow R, de Rinaldis E, Grigoriadis A, Linardopoulos S, Marra P, Tutt ANJ. Integrated genomics and functional validation identifies malignant cell specific dependencies in triple negative breast cancer. Nat Commun 2018; 9:1044. [PMID: 29535384 PMCID: PMC5849766 DOI: 10.1038/s41467-018-03283-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 02/01/2018] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancers (TNBCs) lack recurrent targetable driver mutations but demonstrate frequent copy number aberrations (CNAs). Here, we describe an integrative genomic and RNAi-based approach that identifies and validates gene addictions in TNBCs. CNAs and gene expression alterations are integrated and genes scored for pre-specified target features revealing 130 candidate genes. We test functional dependence on each of these genes using RNAi in breast cancer and non-malignant cells, validating malignant cell selective dependence upon 37 of 130 genes. Further analysis reveals a cluster of 13 TNBC addiction genes frequently co-upregulated that includes genes regulating cell cycle checkpoints, DNA damage response, and malignant cell selective mitotic genes. We validate the mechanism of addiction to a potential drug target: the mitotic kinesin family member C1 (KIFC1/HSET), essential for successful bipolar division of centrosome-amplified malignant cells and develop a potential selection biomarker to identify patients with tumors exhibiting centrosome amplification.
Collapse
Affiliation(s)
- Nirmesh Patel
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Daniel Weekes
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Konstantinos Drosopoulos
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Patrycja Gazinska
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Elodie Noel
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Mamun Rashid
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Hasan Mirza
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
- Cancer Bioinformatics, King's College London, London, SE1 9RT, UK
| | - Jelmar Quist
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
- Cancer Bioinformatics, King's College London, London, SE1 9RT, UK
| | - Fara Brasó-Maristany
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Sumi Mathew
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Riccardo Ferro
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Ana Mendes Pereira
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Cynthia Prince
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Farzana Noor
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Erika Francesch-Domenech
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Rebecca Marlow
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Emanuele de Rinaldis
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
- Precision Immunology Cluster, Sanofi, 640 Memorial Drive, Cambridge, MA, 02149, USA
| | - Anita Grigoriadis
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
- Cancer Bioinformatics, King's College London, London, SE1 9RT, UK
| | - Spiros Linardopoulos
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Pierfrancesco Marra
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK
| | - Andrew N J Tutt
- Breast Cancer Now Research Unit, King's College London, London, SE1 9RT, UK.
- School of Cancer and Pharmaceutical Sciences, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK.
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK.
| |
Collapse
|
6
|
Wu CC, Horowitz DP, Deutsch I, Rahmati R, Schecter JM, Saqi A, Wang TJC. De-escalation of radiation dose for human papillomavirus-positive oropharyngeal head and neck squamous cell carcinoma: A case report and preclinical and clinical literature review. Oncol Lett 2015; 11:141-149. [PMID: 26870181 PMCID: PMC4727039 DOI: 10.3892/ol.2015.3836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 07/28/2015] [Indexed: 12/21/2022] Open
Abstract
Traditionally, head and neck squamous cell carcinoma (HNSCC) has been considered to be a relatively homogeneous disease. However, recent data have demonstrated that human papillomavirus (HPV)-positive and HPV-negative disease are two different clinical entities associated with different outcomes. Preclinical and clinical studies have reported a divergence in treatment strategies as well as prognostic outcomes for HNSCCs that are HPV-positive versus HPV-negative. The present study describes the case of a 52-year-old man who presented with stage IVB cT2N3M0 right tonsillar HPV-positive squamous cell carcinoma. Induction chemotherapy with docetaxel, cisplatin and 5-fluorouracil (TPF), followed by chemoradiation therapy with carboplatin and 70 Gray (Gy) radiation in daily fractions was recommended. The patient completed the TPF and carboplatin treatment; however, he was unable to tolerate the radiation course, receiving a final dose of 46 Gy. A 60-day follow-up right neck salvage dissection was subsequently performed. Despite having received a partial radiation treatment of 46 Gy, the patient had no pathological evidence of disease at 60 days post radiation treatment. Repeat positron emission tomography-computed tomography at 32 months after the right neck dissection revealed no evidence of disease. The present study also discusses the current preclinical in vitro and in vivo targets for HPV-positive HNSCC and the obstacles presented in advancing clinical treatment modalities. Previous preclinical models investigating radiation sensitivity have yielded mixed results. Thus, it is important to understand and establish representative preclinical models for studying HPV and HNSCC to improve clinical research and therapeutic development. This review may guide future understanding of the role of HPV in HNSCC.
Collapse
Affiliation(s)
- Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - David P Horowitz
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Israel Deutsch
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Rahmatullah Rahmati
- Department of Otolaryngology-Head and Neck Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jordan M Schecter
- Department of Medical Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Anjali Saqi
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Tony J C Wang
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
7
|
Rambow F, Job B, Petit V, Gesbert F, Delmas V, Seberg H, Meurice G, Van Otterloo E, Dessen P, Robert C, Gautheret D, Cornell RA, Sarasin A, Larue L. New Functional Signatures for Understanding Melanoma Biology from Tumor Cell Lineage-Specific Analysis. Cell Rep 2015; 13:840-853. [PMID: 26489459 PMCID: PMC5970542 DOI: 10.1016/j.celrep.2015.09.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 05/30/2015] [Accepted: 09/14/2015] [Indexed: 01/08/2023] Open
Abstract
Molecular signatures specific to particular tumor types are required to design treatments for resistant tumors. However, it remains unclear whether tumors and corresponding cell lines used for drug development share such signatures. We developed similarity core analysis (SCA), a universal and unsupervised computational framework for extracting core molecular features common to tumors and cell lines. We applied SCA to mRNA/miRNA expression data from various sources, comparing melanoma cell lines and metastases. The signature obtained was associated with phenotypic characteristics in vitro, and the core genes CAPN3 and TRIM63 were implicated in melanoma cell migration/invasion. About 90% of the melanoma signature genes belong to an intrinsic network of transcription factors governing neural development (TFAP2A, DLX2, ALX1, MITF, PAX3, SOX10, LEF1, and GAS7) and miRNAs (211-5p, 221-3p, and 10a-5p). The SCA signature effectively discriminated between two subpopulations of melanoma patients differing in overall survival, and classified MEKi/BRAFi-resistant and -sensitive melanoma cell lines.
Collapse
Affiliation(s)
- Florian Rambow
- Institut Curie, Normal and Pathological Development of Melanocytes, 91405 Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR3347, 91405 Orsay, France; INSERM U1021, 91405 Orsay, France; Equipe Labellisée - Ligue Nationale contre le Cancer, 91405 Orsay, France
| | - Bastien Job
- Plateforme de Bioinformatique, UMS AMMICA, Gustave-Roussy, 94805 Villejuif, France
| | - Valérie Petit
- Institut Curie, Normal and Pathological Development of Melanocytes, 91405 Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR3347, 91405 Orsay, France; INSERM U1021, 91405 Orsay, France; Equipe Labellisée - Ligue Nationale contre le Cancer, 91405 Orsay, France
| | - Franck Gesbert
- Institut Curie, Normal and Pathological Development of Melanocytes, 91405 Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR3347, 91405 Orsay, France; INSERM U1021, 91405 Orsay, France; Equipe Labellisée - Ligue Nationale contre le Cancer, 91405 Orsay, France
| | - Véronique Delmas
- Institut Curie, Normal and Pathological Development of Melanocytes, 91405 Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR3347, 91405 Orsay, France; INSERM U1021, 91405 Orsay, France; Equipe Labellisée - Ligue Nationale contre le Cancer, 91405 Orsay, France
| | - Hannah Seberg
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Guillaume Meurice
- Plateforme de Bioinformatique, UMS AMMICA, Gustave-Roussy, 94805 Villejuif, France
| | - Eric Van Otterloo
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Philippe Dessen
- Plateforme de Bioinformatique, UMS AMMICA, Gustave-Roussy, 94805 Villejuif, France
| | | | - Daniel Gautheret
- Plateforme de Bioinformatique, UMS AMMICA, Gustave-Roussy, 94805 Villejuif, France
| | - Robert A Cornell
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Alain Sarasin
- Centre National de la Recherche Scientifique (CNRS) UMR8200, Gustave-Roussy and University Paris-Sud, 94805 Villejuif, France
| | - Lionel Larue
- Institut Curie, Normal and Pathological Development of Melanocytes, 91405 Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR3347, 91405 Orsay, France; INSERM U1021, 91405 Orsay, France; Equipe Labellisée - Ligue Nationale contre le Cancer, 91405 Orsay, France.
| |
Collapse
|
8
|
Selection strategies for anticancer antibody discovery: searching off the beaten path. Trends Biotechnol 2015; 33:292-301. [PMID: 25819764 DOI: 10.1016/j.tibtech.2015.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 01/13/2023]
Abstract
Antibody-based drugs represent one of the most successful and promising therapeutic approaches in oncology. Large combinatorial phage antibody libraries are available for the identification of therapeutic antibodies and various technologies exist for their further conversion into multivalent and multispecific formats optimized for the desired pharmacokinetics and the pathological context. However, there is no technology for antigen profiling of intact tumors to identify tumor markers targetable with antibodies. Such constraints have led to a relative paucity of tumor-associated antigens for antibody targeting in oncology. Here we review novel approaches aimed at the identification of antibody-targetable, accessible antigens in intact tumors. We hope that such advanced selection approaches will be useful in the development of next-generation antibody therapies for cancer.
Collapse
|
9
|
Stein AP, Swick AD, Smith MA, Blitzer GC, Yang RZ, Saha S, Harari PM, Lambert PF, Liu CZ, Kimple RJ. Xenograft assessment of predictive biomarkers for standard head and neck cancer therapies. Cancer Med 2015; 4:699-712. [PMID: 25619980 PMCID: PMC4430263 DOI: 10.1002/cam4.387] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/16/2014] [Accepted: 11/18/2014] [Indexed: 12/27/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a challenging cancer to treat with overall 5-year survival on the order of 50–60%. Therefore, predictive biomarkers for this disease would be valuable to provide more effective and individualized therapeutic approaches for these patients. While prognostic biomarkers such as p16 expression correlate with outcome; to date, no predictive biomarkers have been clinically validated for HNSCC. We generated xenografts in immunocompromised mice from six established HNSCC cell lines and evaluated response to cisplatin, cetuximab, and radiation. Tissue microarrays were constructed from pre- and posttreatment tumor samples derived from each xenograft experiment. Quantitative immunohistochemistry was performed using a semiautomated imaging and analysis platform to determine the relative expression of five potential predictive biomarkers: epidermal growth factor receptor (EGFR), phospho-EGFR, phospho-Akt, phospho-ERK, and excision repair cross-complementation group 1 (ERCC1). Biomarker levels were compared between xenografts that were sensitive versus resistant to a specific therapy utilizing a two-sample t-test with equal standard deviations. Indeed the xenografts displayed heterogeneous responses to each treatment, and we linked a number of baseline biomarker levels to response. This included low ERCC1 being associated with cisplatin sensitivity, low phospho-Akt correlated with cetuximab sensitivity, and high total EGFR was related to radiation resistance. Overall, we developed a systematic approach to identifying predictive biomarkers and demonstrated several connections between biomarker levels and treatment response. Despite these promising initial results, this work requires additional preclinical validation, likely involving the use of patient-derived xenografts, prior to moving into the clinical realm for confirmation among patients with HNSCC.
Collapse
Affiliation(s)
- Andrew P Stein
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Adam D Swick
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Molly A Smith
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Grace C Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Robert Z Yang
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Sandeep Saha
- Department of Biostatistics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Paul F Lambert
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Cheng Z Liu
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| |
Collapse
|
10
|
Influence of handling conditions on the establishment and propagation of head and neck cancer patient derived xenografts. PLoS One 2014; 9:e100995. [PMID: 24967635 PMCID: PMC4072729 DOI: 10.1371/journal.pone.0100995] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/02/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Patient derived xenografts (PDXs) for head and neck cancer (HNC) and other cancers represent powerful research platforms. Most groups implant patient tissue into immunodeficient mice immediately although the significance of this time interval is anecdotal. We tested the hypothesis that the time from tumor excision to implantation is crucial for PDX passaging and establishment. METHODS We examined whether time or storage medium affected PDX viability for passaging two established HNC PDXs (UW-SCC34, UW-SCC52). Tumors were harvested, stored in ice-cold media or saline for 0-48 hours, and implanted into new mice. Tumor growth was compared by two-way ANOVA with respect to time and storage condition. Three new HNC PDXs (UW-SCC63-65) were generated by implanting patient tissue into mice immediately (Time 0) and 24 hours after receiving tissue from the operating room. RESULTS Similar quantities of tumor were implanted into each mouse. At the end of the experiment, no significant difference was seen in mean tumor weight between the media and saline storage conditions for UW-SCC34 or UW-SCC52 (p = 0.650 and p = 0.177, respectively). No difference in tumor formation prevalence was seen on the basis of time from harvest to implantation (≥13 of 16 tumors grew at every time point). Histological analysis showed strong similarity to the initial tumor across all groups. Tumors developed at both Time 0 and 24 hours for UW-SCC63 and UW-SCC64. CONCLUSIONS We demonstrated that neither storage medium nor time from tumor excision to implantation (up to 48 hours) affected viability or histological differentiation in a subsequent passage for two HNC PDXs. Moreover, we revealed that fresh patient tissue is viable up to 24 hours post-resection. This information is important as it applies to the development and sharing of PDXs.
Collapse
|
11
|
Huang YY, Hoshino K, Chen P, Wu CH, Lane N, Huebschman M, Liu H, Sokolov K, Uhr JW, Frenkel EP, Zhang X. Immunomagnetic nanoscreening of circulating tumor cells with a motion controlled microfluidic system. Biomed Microdevices 2013; 15:673-681. [PMID: 23109037 PMCID: PMC3584207 DOI: 10.1007/s10544-012-9718-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Combining the power of immunomagnetic assay and microfluidic microchip operations, we successfully detected rare CTCs from clinical blood samples. The microfluidic system is operated in a flip-flop mode, where a computer-controlled rotational holder with an array of microfluidic chips inverts the microchannels. We have demonstrated both theoretically and experimentally that the direction of red blood cell (RBC) sedimentation with regards to the magnetic force required for cell separation is important for capture efficiency, throughput, and purity. The flip-flop operation reduces the stagnation of RBCs and non-specific binding on the capture surface by alternating the direction of the magnetic field with respect to gravity. The developed immunomagnetic microchip-based screening system exhibits high capture rates (more than 90%) for SkBr3, PC3, and Colo205 cell lines in spiked screening experiments and successfully isolates CTCs from patient blood samples. The proposed motion controlled microchip-based immunomagnetic system shows great promise as a clinical tool for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Yu-Yen Huang
- Department of Biomedical Engineering, The University of Texas at Austin, University Station, C0800, Austin, TX, 78712-0238, USA
| | - Kazunori Hoshino
- Department of Biomedical Engineering, The University of Texas at Austin, University Station, C0800, Austin, TX, 78712-0238, USA
| | - Peng Chen
- Department of Biomedical Engineering, The University of Texas at Austin, University Station, C0800, Austin, TX, 78712-0238, USA
| | - Chung-Hsien Wu
- Department of Biomedical Engineering, The University of Texas at Austin, University Station, C0800, Austin, TX, 78712-0238, USA
| | - Nancy Lane
- Harold C. Simons Comprehensive Cancer Center of the University of Texas Southwestern Medical Center, USA, 5323 Harry Hines Boulevard Dallas, TX, 7390-852, USA
| | - Michael Huebschman
- Harold C. Simons Comprehensive Cancer Center of the University of Texas Southwestern Medical Center, USA, 5323 Harry Hines Boulevard Dallas, TX, 7390-852, USA
| | - Huaying Liu
- Harold C. Simons Comprehensive Cancer Center of the University of Texas Southwestern Medical Center, USA, 5323 Harry Hines Boulevard Dallas, TX, 7390-852, USA
| | - Konstantin Sokolov
- Department of Imaging Physics, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Jonathan W. Uhr
- Harold C. Simons Comprehensive Cancer Center of the University of Texas Southwestern Medical Center, USA, 5323 Harry Hines Boulevard Dallas, TX, 7390-852, USA
| | - Eugene P. Frenkel
- Harold C. Simons Comprehensive Cancer Center of the University of Texas Southwestern Medical Center, USA, 5323 Harry Hines Boulevard Dallas, TX, 7390-852, USA
| | - Xiaojing Zhang
- Department of Biomedical Engineering, The University of Texas at Austin, University Station, C0800, Austin, TX, 78712-0238, USA
| |
Collapse
|
12
|
Kobold S, Völk S, Clauditz T, Küpper NJ, Minner S, Tufman A, Düwell P, Lindner M, Koch I, Heidegger S, Rothenfuer S, Schnurr M, Huber RM, Wilczak W, Endres S. Interleukin-22 is frequently expressed in small- and large-cell lung cancer and promotes growth in chemotherapy-resistant cancer cells. J Thorac Oncol 2013; 8:1032-42. [PMID: 23774470 DOI: 10.1097/jto.0b013e31829923c8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION In lung cancer, interleukin-22 (IL-22) expression within primary tissue has been demonstrated, but the frequency and the functional consequence of IL-22 signaling have not been addressed. This study aims at analyzing the cellular effects of IL-22 on lung carcinoma cell lines and the prognostic impact of IL-22 tissue expression in lung cancer patients. METHODS Biological effects of IL-22 signaling were investigated in seven lung cancer cell lines by Western blot, flow cytometry, real-time polymerase chain reaction, and proliferation assays. Tumor tissue specimens of two cohorts with a total of 2300 lung cancer patients were tested for IL-22 expression by immunohistochemistry. IL-22 serum concentrations were analyzed in 103 additional patients by enzyme-linked immunosorbent assay. RESULTS We found the IL-22 receptor 1 (IL-22-R1) to be expressed in six of seven lung cancer cell lines. However IL-22 signaling was functional in only four cell lines, where IL-22 induced signal transducer activator of transcription 3 phosphorylation and increased cell proliferation. Furthermore, IL-22 induced the expression of antiapoptotic B-cell lymphoma 2, but did not rescue tumor cells from carboplatin-induced apoptosis. Cisplatin-resistant cell lines showed a significant up-regulation of IL-22-R1 along with a stronger proliferative response to IL-22 stimulation. IL-22 was preferentially expressed in small- and large-cell lung carcinoma (58% and 46% of cases, respectively). However, no correlation between IL-22 expression by immunohistochemistry and prognosis was observed. CONCLUSION IL-22 is frequently expressed in lung cancer tissue. Enhanced IL-22-R1 expression and signaling in chemotherapy-refractory cell lines are indicative of a protumorigenic function of IL-22 and may contribute to a more aggressive phenotype.
Collapse
Affiliation(s)
- Sebastian Kobold
- Department of Internal Medicine IV, Division of Clinical Pharmacology and Center of Integrated Protein Science, Ludwig-Maximilians Universität München, Member of the German Center for Lung Research, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kimple RJ, Harari PM, Torres AD, Yang RZ, Soriano BJ, Yu M, Armstrong EA, Blitzer GC, Smith MA, Lorenz LD, Lee D, Yang DT, McCulloch TM, Hartig GK, Lambert PF. Development and characterization of HPV-positive and HPV-negative head and neck squamous cell carcinoma tumorgrafts. Clin Cancer Res 2012; 19:855-64. [PMID: 23251001 DOI: 10.1158/1078-0432.ccr-12-2746] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE To develop a clinically relevant model system to study head and neck squamous cell carcinoma (HNSCC), we have established and characterized a direct-from-patient tumorgraft model of human papillomavirus (HPV)-positive and HPV-negative cancers. EXPERIMENTAL DESIGN Patients with newly diagnosed or recurrent HNSCC were consented for donation of tumor specimens. Surgically obtained tissue was implanted subcutaneously into immunodeficient mice. During subsequent passages, both formalin-fixed/paraffin-embedded as well as flash-frozen tissues were harvested. Tumors were analyzed for a variety of relevant tumor markers. Tumor growth rates and response to radiation, cisplatin, or cetuximab were assessed and early passage cell strains were developed for rapid testing of drug sensitivity. RESULTS Tumorgrafts have been established in 22 of 26 patients to date. Significant diversity in tumorgraft tumor differentiation was observed with good agreement in degree of differentiation between patient tumor and tumorgraft (Kappa 0.72). Six tumorgrafts were HPV-positive on the basis of p16 staining. A strong inverse correlation between tumorgraft p16 and p53 or Rb was identified (Spearman correlations P = 0.085 and P = 0.002, respectively). Significant growth inhibition of representative tumorgrafts was shown with cisplatin, cetuximab, or radiation treatment delivered over a two-week period. Early passage cell strains showed high consistency in response to cancer therapy between tumorgraft and cell strain. CONCLUSIONS We have established a robust human tumorgraft model system for investigating HPV-positive and HPV-negative HNSCC. These tumorgrafts show strong correlation with the original tumor specimens and provide a powerful resource for investigating mechanisms of therapeutic response as well as preclinical testing.
Collapse
Affiliation(s)
- Randall J Kimple
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lazar DC, Cho EH, Luttgen MS, Metzner TJ, Uson ML, Torrey M, Gross ME, Kuhn P. Cytometric comparisons between circulating tumor cells from prostate cancer patients and the prostate-tumor-derived LNCaP cell line. Phys Biol 2012; 9:016002. [PMID: 22306736 DOI: 10.1088/1478-3975/9/1/016002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Many important experiments in cancer research are initiated with cell line data analysis due to the ease of accessibility and utilization. Recently, the ability to capture and characterize circulating tumor cells (CTCs) has become more prevalent in the research setting. This ability to detect, isolate and analyze CTCs allows us to directly compare specific protein expression levels found in patient CTCs to cell lines. In this study, we use immunocytochemistry to compare the protein expression levels of total cytokeratin (CK) and androgen receptor (AR) in CTCs and cell lines from patients with prostate cancer to determine what translational insights might be gained through the use of cell line data. A non-enrichment CTC detection assay enables us to compare cytometric features and relative expression levels of CK and AR by indirect immunofluorescence from prostate cancer patients against the prostate cancer cell line LNCaP. We measured physical characteristics of these two groups and observed significant differences in cell size, fluorescence intensity and nuclear to cytoplasmic ratio. We hope that these experiments will initiate a foundation to allow cell line data to be compared against characteristics of primary cells from patients.
Collapse
Affiliation(s)
- Daniel C Lazar
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, GAC-1200, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | |
Collapse
|