1
|
Kalinin AA, Arevalo J, Serrano E, Vulliard L, Tsang H, Bornholdt M, Muñoz AF, Sivagurunathan S, Rajwa B, Carpenter AE, Way GP, Singh S. A versatile information retrieval framework for evaluating profile strength and similarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.01.587631. [PMID: 38617315 PMCID: PMC11014546 DOI: 10.1101/2024.04.01.587631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
In profiling assays, thousands of biological properties are measured across many samples, yielding biological discoveries by capturing the state of a cell population, often at the single-cell level. However, for profiling datasets, it has been challenging to evaluate the phenotypic activity of a sample and the phenotypic consistency among samples, due to profiles' high dimensionality, heterogeneous nature, and non-linear properties. Existing methods leave researchers uncertain where to draw boundaries between meaningful biological response and technical noise. Here, we developed a statistical framework that uses the well-established mean average precision (mAP) as a single, data-driven metric to bridge this gap. We validated the mAP framework against established metrics through simulations and real-world data applications, revealing its ability to capture subtle and meaningful biological differences in cell state. Specifically, we used mAP to assess both phenotypic activity for a given perturbation (or a sample) as well as consistency within groups of perturbations (or samples) across diverse high-dimensional datasets. We evaluated the framework on different profile types (image, protein, and mRNA profiles), perturbation types (CRISPR gene editing, gene overexpression, and small molecules), and profile resolutions (single-cell and bulk). Our open-source software allows this framework to be applied to identify interesting biological phenomena and promising therapeutics from large-scale profiling data.
Collapse
Affiliation(s)
| | - John Arevalo
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| | - Erik Serrano
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora CO, USA
| | - Loan Vulliard
- Systems Immunology and Single-Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hillary Tsang
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| | - Michael Bornholdt
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| | - Alán F. Muñoz
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| | | | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette IN, USA
| | - Anne E. Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| | - Gregory P. Way
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora CO, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge MA, USA
| |
Collapse
|
2
|
Mishra S, Kumarasamy M. Microfluidics engineering towards personalized oncology-a review. IN VITRO MODELS 2023; 2:69-81. [PMID: 39871996 PMCID: PMC11756504 DOI: 10.1007/s44164-023-00054-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 01/29/2025]
Abstract
Identifying and monitoring the presence of cancer metastasis and highlighting inter-and intratumoral heterogeneity is a central tenet of targeted precision oncology medicine (POM). This process of relocation of cancer cells is often referred to as the missing link between a tumor and metastasis. In recent years, microfluidic technologies have been developed to isolate a plethora of different biomarkers, such as circulating tumor cells (CTCs), tumor-derived vesicles (exosomes), or cell/free nucleic acids and proteins directly from patients' blood samples. With the advent of microfluidic developments, minimally invasive and quantitative assessment of different tumors is becoming a reality. This short review article will touch briefly on how microfluidics at early-stage achievements can be combined or developed with the active vs passive microfluidic technologies, depending on whether they utilize external fields and forces (active) or just microchannel geometry and inherent fluid forces (passive) from the market to precision oncology research and our future prospectives in terms of the emergence of ultralow cost and rapid prototyping of microfluidics in precision oncology.
Collapse
Affiliation(s)
- Sushmita Mishra
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur (NIPERHajipur) Export Promotion Industrial Park (EPIP), Industrial Area, Vaishali, 844102 Bihar India
| | - Murali Kumarasamy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur (NIPERHajipur) Export Promotion Industrial Park (EPIP), Industrial Area, Vaishali, 844102 Bihar India
| |
Collapse
|
3
|
Development of Prognostic Features of Hepatocellular Carcinoma Based on Metabolic Gene Classification and Immune and Oxidative Stress Characteristic Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:1847700. [PMID: 36860731 PMCID: PMC9969974 DOI: 10.1155/2023/1847700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/18/2022] [Accepted: 11/24/2022] [Indexed: 02/20/2023]
Abstract
Background The molecular classification of HCC premised on metabolic genes might give assistance for diagnosis, therapy, prognosis prediction, immune infiltration, and oxidative stress in addition to supplementing the limitations of the clinical staging system. This would help to better represent the deeper features of HCC. Methods TCGA datasets combined with GSE14520 and HCCDB18 datasets were used to determine the metabolic subtype (MC) using ConsensusClusterPlus. ssGSEA method was used to calculate the IFNγ score, the oxidative stress pathway scores, and the score distribution of 22 distinct immune cells, and their differential expressions were assessed with the use of CIBERSORT. To generate a subtype classification feature index, LDA was utilized. Screening of the metabolic gene coexpression modules was done with the help of WGCNA. Results Three MCs (MC1, MC2, and MC3) were identified and showed different prognoses (MC2-poor and MC1-better). Although MC2 had a high immune microenvironment infiltration, T cell exhaustion markers were expressed at a high level in MC2 in contrast with MC1. Most oxidative stress-related pathways are inhibited in the MC2 subtype and activated in the MC1 subtype. The immunophenotyping of pan-cancer showed that the C1 and C2 subtypes with poor prognosis accounted for significantly higher proportions of MC2 and MC3 subtypes than MC1, while the better prognostic C3 subtype accounted for significantly lower proportions of MC2 than MC1. As per the findings of the TIDE analysis, MC1 had a greater likelihood of benefiting from immunotherapeutic regimens. MC2 was found to have a greater sensitivity to traditional chemotherapy drugs. Finally, 7 potential gene markers indicate HCC prognosis. Conclusion The difference (variation) in tumor microenvironment and oxidative stress among metabolic subtypes of HCC was compared from multiple angles and levels. A complete and thorough clarification of the molecular pathological properties of HCC, the exploration of reliable markers for diagnosis, the improvement of the cancer staging system, and the guiding of individualized treatment of HCC all gain benefit greatly from molecular classification associated with metabolism.
Collapse
|
4
|
Applications of mass spectroscopy in understanding cancer proteomics. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
5
|
Chen R, Wang X, Deng X, Chen L, Liu Z, Li D. CPDR: An R Package of Recommending Personalized Drugs for Cancer Patients by Reversing the Individual’s Disease-Related Signature. Front Pharmacol 2022; 13:904909. [PMID: 35795573 PMCID: PMC9252520 DOI: 10.3389/fphar.2022.904909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Due to cancer heterogeneity, only some patients can benefit from drug therapy. The personalized drug usage is important for improving the treatment response rate of cancer patients. The value of the transcriptome of patients has been recently demonstrated in guiding personalized drug use, and the Connectivity Map (CMAP) is a reliable computational approach for drug recommendation. However, there is still no personalized drug recommendation tool based on transcriptomic profiles of patients and CMAP. To fill this gap, here, we proposed such a feasible workflow and a user-friendly R package—Cancer-Personalized Drug Recommendation (CPDR). CPDR has three features. 1) It identifies the individual disease signature by using the patient subgroup with transcriptomic profiles similar to those of the input patient. 2) Transcriptomic profile purification is supported for the subgroup with high infiltration of non-cancerous cells. 3) It supports in silico drug efficacy assessment using drug sensitivity data on cancer cell lines. We demonstrated the workflow of CPDR with the aid of a colorectal cancer dataset from GEO and performed the in silico validation of drug efficacy. We further assessed the performance of CPDR by a pancreatic cancer dataset with clinical response to gemcitabine. The results showed that CPDR can recommend promising therapeutic agents for the individual patient. The CPDR R package is available at https://github.com/AllenSpike/CPDR.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong Li
- *Correspondence: Zhongyang Liu, ; Dong Li,
| |
Collapse
|
6
|
Point-of-care detection assay based on biomarker-imprinted polymer for different cancers: a state-of-the-art review. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04085-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
7
|
Amiri-Dashatan N, Koushki M, Rezaei-Tavirani M. Mass Spectrometry-Based Proteomics Research to Fight COVID-19: An Expert Review on Hopes and Challenges. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:19-34. [PMID: 35005991 DOI: 10.1089/omi.2021.0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome (SARS)-CoV-2 infection is a systemic disease and a major planetary health burden. While SARS-CoV-2 impacts host biology extensively, our knowledge of these alterations from a systems perspective remains incomplete. Moreover, there is currently only a limited description of this systemic disease. For precision diagnosis and treatment of SARS-CoV-2, multiomics technologies and systems science research offer significant prospects. This expert review offers a critical analysis of the prospects and challenges of the emerging mass spectrometry-based proteomics approaches to the study of COVID-19 as seen through a systems medicine lens. We also discuss the ways in which proteomics is poised to offer hope for diagnostics and therapeutics innovation on SARS-CoV-2 infection as the disease transitions from a pandemic to an endemic disease, and thus further challenging the health systems and services worldwide in the coming decade. Proteomics is an important high-throughput technology platform to achieve a functional overview of the ways in which COVID-19 changes host biology, and hence, can help identify possible points of entry for innovation in medicines and vaccines, among others.
Collapse
Affiliation(s)
- Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
8
|
Abstract
Hepatoblastoma (HB) is the predominant primary liver tumor in children. While the prognosis is favorable when the tumor can be resected, the outcome is dismal for patients with progressed HB. Therefore, a better understanding of the molecular mechanisms responsible for HB is imperative for early detection and effective treatment. Sequencing analysis of human HB specimens unraveled the pivotal role of Wnt/β-catenin pathway activation in this disease. Nonetheless, β-catenin activation alone does not suffice to induce HB, implying the need for additional alterations. Perturbations of several pathways, including Hippo, Hedgehog, NRF2/KEAP1, HGF/c-Met, NK-1R/SP, and PI3K/AKT/mTOR cascades and aberrant activation of c-MYC, n-MYC, and EZH2 proto-oncogenes, have been identified in HB, although their role requires additional investigation. Here, we summarize the current knowledge on HB molecular pathogenesis, the relevance of the preclinical findings for the human disease, and the innovative therapeutic strategies that could be beneficial for the treatment of HB patients.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Antonio Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefano Cairo
- XenTech, Evry, France,Istituto di Ricerca Pediatrica, Padova, Italy
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
9
|
Gil-Bazo I. Oncogenes in Cancer: Using the Problem as Part of the Solution. Cancers (Basel) 2020; 12:cancers12113373. [PMID: 33202595 PMCID: PMC7696071 DOI: 10.3390/cancers12113373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022] Open
Abstract
Human cancer is considered to have a multifactorial origin [...].
Collapse
Affiliation(s)
- Ignacio Gil-Bazo
- Department of Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; ; Tel.: +34-948-255400; Fax: +34-948-255500
- Program of Solid Tumors, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
10
|
Melaiu O, Lucarini V, Giovannoni R, Fruci D, Gemignani F. News on immune checkpoint inhibitors as immunotherapy strategies in adult and pediatric solid tumors. Semin Cancer Biol 2020; 79:18-43. [PMID: 32659257 DOI: 10.1016/j.semcancer.2020.07.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/19/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have shown unprecedented benefits in various adult cancers, and this success has prompted the exploration of ICI therapy even in childhood malignances. Although the use of ICIs as individual agents has achieved disappointing response rates, combinational therapies are likely to promise better results. However, only a subset of patients experienced prolonged clinical effects, thus suggesting the need to identify robust bio-markers that predict individual clinical response or resistance to ICI therapy as the main challenge. In this review, we focus on how the use of ICIs in adult cancers can be translated into pediatric malignances. We discuss the physiological mechanism of action of each IC, including PD-1, PD-L1 and CTLA-4 and the new emerging ones, LAG-3, TIM-3, TIGIT, B7-H3, BTLA and IDO-1, and evaluate their prognostic value in both adult and childhood tumors. Furthermore, we offer an overview of preclinical models and clinical trials currently under investigation to improve the effectiveness of cancer immunotherapies in these patients. Finally, we outline the main predictive factors that influence the efficacy of ICIs, in order to lay the basis for the development of a pan-cancer immunogenomic model, able to direct young patients towards more specific immunotherapy.
Collapse
Affiliation(s)
- Ombretta Melaiu
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Valeria Lucarini
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Doriana Fruci
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy.
| | | |
Collapse
|
11
|
Al-Eitan LN, Rababa'h DM, Alghamdi MA, Khasawneh RH. The influence of an IL-4 variable number tandem repeat (VNTR) polymorphism on breast cancer susceptibility. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:201-207. [PMID: 31692576 PMCID: PMC6716593 DOI: 10.2147/pgpm.s220571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/06/2019] [Indexed: 01/20/2023]
Abstract
Backgrounds Breast cancer (BC) is one of the most widespread cancers globally. Understanding the etiology of BC may help in determining the various risk factors involved in its malignancy. Certain genetic mutations are considered to play a key role in increasing the risk of BC. Objectives In this study, we explored the correlation between a variable number tandem repeat (VNTR) polymorphism in the IL-4 gene and BC. Methods PCR and subsequent gel electrophoresis were used to genotype this variant in 360 Jordanian women (180 BC patients and 180 controls). In addition, phenotype–genotype analysis was carried out. Results Our findings illustrate that there is no significant relationship between the variant genotypes in the IL-4 gene and BC among Jordanian females. Other than body mass index and tumor differentiation (p< 0.05), none of the clinical and pathological parameters of BC patients exhibited any association with the variant genotypes. Conclusions From this study, we propose that the IL-4 genetic variant does not impact BC development and progression but that it could influence the disease prognosis.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan.,Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Doaa M Rababa'h
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mansour A Alghamdi
- Anatomy Department, Faculty of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Rame H Khasawneh
- Department of Hematopathology, King Hussein Medical Center (KHMC), Jordanian Royal Medical Services (RMS), Amman 11118, Jordan
| |
Collapse
|
12
|
Liu H, Ye Z, Wang X, Wei L, Xiao L. Molecular and living cell dynamic assays with optical microscopy imaging techniques. Analyst 2019; 144:859-871. [PMID: 30444498 DOI: 10.1039/c8an01420e] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Generally, the message elucidated by the conventional analytical methods overlooks the heterogeneity of single objects, where the behavior of individual molecules is shielded. With the advent of optical microscopy imaging techniques, it is possible to identify, visualize and track individual molecules or nanoparticles under a biological environment with high temporal and spatial resolution. In this work, we summarize the commonly adopted optical microscopy techniques for bio-analytical assays in living cells, including total internal reflection fluorescence microscopy (TIRFM), super-resolution optical microscopy (SRM), and dark-field optical microscopy (DFM). The basic principles of these methods and some recent interesting applications in molecular detection and single-particle tracking are introduced. Moreover, the development in high-dimensional optical microscopy to achieve three-dimensional (3-D) as well as sub-diffraction localization and tracking of biomolecules is also highlighted.
Collapse
Affiliation(s)
- Hua Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin, 300071, China.
| | | | | | | | | |
Collapse
|
13
|
Petricoin E, Wulfkuhle J, Howard M, Pierobon M, Espina V, Luchini A, Liotta LA. RPPA: Origins, Transition to a Validated Clinical Research Tool, and Next Generations of the Technology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1188:1-19. [PMID: 31820380 DOI: 10.1007/978-981-32-9755-5_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RPPA technology has graduated from a research tool to an essential component of clinical drug discovery research and personalized medicine. Next generations of RPPA technology will be a single clinical instrument that integrates all the steps of the workflow.
Collapse
Affiliation(s)
- Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Julie Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Marissa Howard
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Marielena Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Alessandra Luchini
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA.
| |
Collapse
|
14
|
Xiao H, Xu D, Chen P, Zeng G, Wang X, Zhang X. Identification of Five Genes as a Potential Biomarker for Predicting Progress and Prognosis in Adrenocortical Carcinoma. J Cancer 2018; 9:4484-4495. [PMID: 30519354 PMCID: PMC6277665 DOI: 10.7150/jca.26698] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Adrenocortical carcinoma (ACC) is a limited endocrine fatality with a minor diagnosis and rare remedial options. The progressive and predictive meaning of message RNA (mRNA) expression oddity in ACC has been studied extensively in recent years. However, differences in measurement platforms and lab protocols as well as small sample sizes can render gene expression levels incomparable. Methods: An extensive study of GEO datasets was conducted to define potential mRNA biomarkers for ACC. The study compared the mRNA expression profiles of ACC tissues and neighboring noncancerous adrenal tissues in the pair. The study covered a sum of 165 tumors and 36 benign control samples. Hub genes were identified through a protein-protein interaction (PPI) network and Robust Rank Aggregation method. Then the Cancer Genome Atlas (TCGA) and Oncomine database were used to perform the validation of hub genes. 4 ACC tissues and 4 normal tissues were collected and then Polymerase Chain Reaction (PCR), Western-blot and immunofluorescence were conducted to validate the expression of five hub genes. Results: We identified five statistically significant genes (TOP2A, NDC80, CEP55, CDKN3, CDK1) corrected with clinical features. The expression of five hub genes in TCGA and Oncomine database were significantly overexpressed in ACC compared with normal ones. Among all the TCGA ACC cases, the strong expression of TOP2A (logrank p=1.4e-04, HR=4.7), NDC80 (logrank p=8.8e-05, HR=4.9), CEP55 (logrank p=5.2e-07, HR=8.6), CDKN3 (log rank p=2.3e-06, HR=7.6) and CDK1 (logrank p=7e-08, HR=11) were correlated with low comprehensive survival, disease free survival (logrank p < 0.001), pathology stage and pathology T stage (FDR < 0.001). PCR results showed that the transcriptional levels of these five genes were significantly higher in ACC tissues than in normal tissues. The western blotting results also showed that the translational level of TOP2A was significantly higher in tumor tissues than in normal tissues. The results of immunofluorescence showed that TOP2A was abundantly observed in the adrenal cortical cell membrane and nucleus and its expression in ACC tissues was significantly higher than that in normal tissues. Conclusions: The distinguished five genes may be utilized to form a board of progressive and predictive biomarkers for ACC for clinical purpose.
Collapse
Affiliation(s)
- He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Deqiang Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Guang Zeng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China.,Biomedical Engineering, Stony Brook University, New York 11790
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
15
|
Campbell JM, Balhoff JB, Landwehr GM, Rahman SM, Vaithiyanathan M, Melvin AT. Microfluidic and Paper-Based Devices for Disease Detection and Diagnostic Research. Int J Mol Sci 2018; 19:E2731. [PMID: 30213089 PMCID: PMC6164778 DOI: 10.3390/ijms19092731] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in microfluidic devices, nanoparticle chemistry, fluorescent microscopy, and biochemical techniques such as genetic identification and antibody capture have provided easier and more sensitive platforms for detecting and diagnosing diseases as well as providing new fundamental insight into disease progression. These advancements have led to the development of new technology and assays capable of easy and early detection of pathogenicity as well as the enhancement of the drug discovery and development pipeline. While some studies have focused on treatment, many of these technologies have found initial success in laboratories as a precursor for clinical applications. This review highlights the current and future progress of microfluidic techniques geared toward the timely and inexpensive diagnosis of disease including technologies aimed at high-throughput single cell analysis for drug development. It also summarizes novel microfluidic approaches to characterize fundamental cellular behavior and heterogeneity.
Collapse
Affiliation(s)
- Joshua M Campbell
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Joseph B Balhoff
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Grant M Landwehr
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Sharif M Rahman
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | | - Adam T Melvin
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
16
|
Patterns and Determinants of Attitudes towards Genetic Risk of Cancer: Case Study in a Malaysian Public University. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4682431. [PMID: 30112391 PMCID: PMC6077651 DOI: 10.1155/2018/4682431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/29/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022]
Abstract
Genetic risk to cancer is a knowledge largely confined to experts and the more educated sectors of the developed western countries. The perception of genetic susceptibility to cancer among the masses is fragmented, particularly in developing countries. As cancer diseases affect developing countries as much as developed nations, it is imperative to study perception and reception of genetic risk to cancer in Southeast Asia. Here, we report on a novel case study to gauge the awareness and attitudes towards genetic determination of cancer among the undergraduates of a Malaysian public university. A total of 272 university undergraduate students completed an online questionnaire. On causes of cancer, the respondents believed that cancer is caused by lifestyle and environmental factors, but those with science background were more likely to associate it with genetic factors. The results on awareness of genetic profiling of cancer risk showed that there are significant differences between those with science and nonscience background but there are no significant differences for gender and socioeconomic background. As for attitudes towards cancer risk, female respondents, those from middle socioeconomic status and science background, are more likely to believe in genetic determinism of cancer. The findings have implications on target population segmentation in strategic health communication on cancer.
Collapse
|
17
|
Rye MB, Bertilsson H, Andersen MK, Rise K, Bathen TF, Drabløs F, Tessem MB. Cholesterol synthesis pathway genes in prostate cancer are transcriptionally downregulated when tissue confounding is minimized. BMC Cancer 2018; 18:478. [PMID: 29703166 PMCID: PMC5922022 DOI: 10.1186/s12885-018-4373-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 04/15/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The relationship between cholesterol and prostate cancer has been extensively studied for decades, where high levels of cellular cholesterol are generally associated with cancer progression and less favorable outcomes. However, the role of in vivo cellular cholesterol synthesis in this process is unclear, and data on the transcriptional activity of cholesterol synthesis pathway genes in tissue from prostate cancer patients are inconsistent. METHODS A common problem with cancer tissue data from patient cohorts is the presence of heterogeneous tissue which confounds molecular analysis of the samples. In this study we present a general method to minimize systematic confounding from stroma tissue in any prostate cancer cohort comparing prostate cancer and normal samples. In particular we use samples assessed by histopathology to identify genes enriched and depleted in prostate stroma. These genes are then used to assess stroma content in tissue samples from other prostate cancer cohorts where no histopathology is available. Differential expression analysis is performed by comparing cancer and normal samples where the average stroma content has been balanced between the sample groups. In total we analyzed seven patient cohorts with prostate cancer consisting of 1713 prostate cancer and 230 normal tissue samples. RESULTS When stroma confounding was minimized, differential gene expression analysis over all cohorts showed robust and consistent downregulation of nearly all genes in the cholesterol synthesis pathway. Additional Gene Ontology analysis also identified cholesterol synthesis as the most significantly altered metabolic pathway in prostate cancer at the transcriptional level. CONCLUSION The surprising observation that cholesterol synthesis genes are downregulated in prostate cancer is important for our understanding of how prostate cancer cells regulate cholesterol levels in vivo. Moreover, we show that tissue heterogeneity explains the lack of consistency in previous expression analysis of cholesterol synthesis genes in prostate cancer.
Collapse
Affiliation(s)
- Morten Beck Rye
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, P.O. Box 8905, NO-7491 Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Helena Bertilsson
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, P.O. Box 8905, NO-7491 Trondheim, Norway
- Department of Urology, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Maria K. Andersen
- MI Lab, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Kjersti Rise
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, P.O. Box 8905, NO-7491 Trondheim, Norway
| | - Tone F. Bathen
- MI Lab, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Finn Drabløs
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, P.O. Box 8905, NO-7491 Trondheim, Norway
| | - May-Britt Tessem
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- MI Lab, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| |
Collapse
|
18
|
Graybill RM, Cardenosa-Rubio MC, Yang H, Johnson MD, Bailey RC. Multiplexed microRNA Expression Profiling by Combined Asymmetric PCR and Label-Free Detection using Silicon Photonic Sensor Arrays. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2018; 10:1618-1623. [PMID: 30275912 PMCID: PMC6162071 DOI: 10.1039/c8ay00190a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Analysis methods based upon the quantitative, real-time polymerase chain reaction are extremely powerful; however, they face intrinsic limitations in terms of target multiplexing. In contrast, silicon photonic microring resonators represent a modularly multiplexable sensor array technology that is well-suited to the analysis of targeted biomarker panels. In this manuscript we employ an asymmetric polymerase chain reaction approach to selectively amplify copies of cDNAs generated from targeted miRNAs before multiplexed, label-free quantitation through hybridization to microring resonator arrays pre-functionalized with capture sequences. This method, which shows applicability to low input amounts and a large dynamic range, was demonstrated for the simultaneous detection of eight microRNA targets from twenty primary brain tumor samples with expression profiles in good agreement with literature precedent.
Collapse
Affiliation(s)
- Richard M. Graybill
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Matthews Ave., Urbana, IL 61801, USA
| | - Maria C. Cardenosa-Rubio
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Matthews Ave., Urbana, IL 61801, USA
- Department of Chemistry, University of Michigan, 930 N. University Ave. Ann Arbor, MI 48104, USA
| | - Hongwei Yang
- Department of Neurological Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02215, USA
- Department of Neurological Surgery, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - Mark D. Johnson
- Department of Neurological Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02215, USA
- Department of Neurological Surgery, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - Ryan C. Bailey
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Matthews Ave., Urbana, IL 61801, USA
- Department of Chemistry, University of Michigan, 930 N. University Ave. Ann Arbor, MI 48104, USA
| |
Collapse
|
19
|
Sorbello V, Fuso L, Sfiligoi C, Scafoglio C, Ponzone R, Biglia N, Weisz A, Sismondi P, De Bortoli M. Quantitative Real-Time RT-PCR Analysis of Eight Novel Estrogen-Regulated Genes in Breast Cancer. Int J Biol Markers 2018; 18:123-9. [PMID: 12841681 DOI: 10.1177/172460080301800205] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Biological markers capable of predicting the risk of recurrence and the response to treatment in breast cancer are eagerly awaited. Estrogen and progesterone receptors (ER, PgR) in tumor cells mark cancers that are more likely to respond to endocrine treatment, but up to 40% of such patients do not respond. Here, the expression of a group of estrogen-regulated genes, previously identified by microarray analysis of in vitro models, was measured in breast tumors and possible associations with other clinicopathological variables were investigated. Methods The expression of CD24, CD44, HAT-1, BAK-1, G1P3, TIEG, NRP-1 and RXRα was measured by quantitative real-time RT-PCR on RNA from eighteen primary breast tumors. Statistical analyses were used to identify correlations among the eight genes and the available clinicopathological data. Results Variable expression levels of all the genes were observed in all the samples examined. Significant associations of CD24 with tumor size, CD44 with lymph node invasion, and HAT-1 and BAK-1 with ER positivity were found. The possible combinatorial value of these genes was assessed. Unsupervised hierarchical clustering analysis demonstrated that the expression profile of these genes was able to predict ER status with an acceptable approximation. Conclusions Eight novel potential markers for breast cancer have been preliminarily characterized. As expected from in vitro data, their expression is able to discriminate ER- versus ER+ tumors.
Collapse
Affiliation(s)
- V Sorbello
- Laboratory of Oncological Gynecology, Institute for Cancer Research and Treatment, Candiolo, Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Voong KR, Feliciano J, Becker D, Levy B. Beyond PD-L1 testing-emerging biomarkers for immunotherapy in non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:376. [PMID: 29057236 PMCID: PMC5635257 DOI: 10.21037/atm.2017.06.48] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/08/2017] [Indexed: 12/26/2022]
Abstract
Recently, a firmer understanding of tumor immunology and tumor escape mechanisms has led to the development of immune checkpoint inhibitors, antibodies against programmed death-1 (PD-1) and its ligand (PD-L1). Nivolumab, pembrolizumab, and atezolizumab have dramatically altered the treatment paradigm in non-small cell lung cancer (NSCLC) and have each demonstrated improvements in outcomes and quality of life when compared to chemotherapy. Enrichment strategies to better select those patients more likely to respond have identified PD-L1 staining by immunohistochemistry (IHC) to be a predictive biomarker in both treatment naïve and refractory patients. Unfortunately, many challenges exist with this strategy and underscore the need for further exploration for more reliable biomarkers. Multiple tissue and plasma-based enrichment strategies have been identified in the hope of identifying patients more likely to benefit from checkpoint inhibitors. These include tumor mutational load; the "inflamed phenotype" including tumor infiltrating lymphocytes (TILS) and immunoscore; T-cell receptor clonality; gene signatures, and several plasma biomarkers. Several studies have revealed many of these biomarkers to be reliable predictors of response to immune checkpoint inhibitors across multiple tumor types. Given the small nature of these studies, additional prospective studies are warranted to formalize and validate each of these enrichment strategies.
Collapse
Affiliation(s)
- Khinh Ranh Voong
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Josephine Feliciano
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Becker
- Langone Cancer Center, Veterans Association Hospital, New York University, New York, NY, USA
| | - Benjamin Levy
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Sibley Memorial Hospital, Washington, DC, USA
| |
Collapse
|
21
|
Tadimety A, Syed A, Nie Y, Long CR, Kready KM, Zhang JXJ. Liquid biopsy on chip: a paradigm shift towards the understanding of cancer metastasis. Integr Biol (Camb) 2017; 9:22-49. [DOI: 10.1039/c6ib00202a] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Amogha Tadimety
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
| | - Abeer Syed
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
| | - Yuan Nie
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
| | - Christina R. Long
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
| | - Kasia M. Kready
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
| | - John X. J. Zhang
- Thayer School of Engineering at Dartmouth College, Hanover NH, 03755, USA
- Dartmouth-Hitchcock Norris Cotton Cancer Center, Lebanon NH, 03766, USA
| |
Collapse
|
22
|
Tekin N, Omidvar N, Morris TP, Conget P, Bruna F, Timar B, Gagyi E, Basak R, Naik O, Auewarakul C, Sritana N, Levy D, Cerci JJ, Bydlowski SP, Pereira J, Dimamay MP, Natividad F, Chung JK, Belder N, Kuzu I, Paez D, Dondi M, Carr R, Ozdag H, Padua RA. Protocol for qRT-PCR analysis from formalin fixed paraffin embedded tissue sections from diffuse large b-cell lymphoma: Validation of the six-gene predictor score. Oncotarget 2016; 7:83319-83329. [PMID: 27825111 PMCID: PMC5347772 DOI: 10.18632/oncotarget.13066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 09/24/2016] [Indexed: 11/28/2022] Open
Abstract
As a part of an international study on the molecular analysis of Diffuse Large B-cell Lymphoma (DLBCL), a robust protocol for gene expression analysis from RNA extraction to qRT-PCR using Formalin Fixed Paraffin Embedded tissues was developed. Here a study was conducted to define a strategy to validate the previously reported 6-gene (LMO2, BCL6, FN1, CCND2, SCYA3 and BCL2) model as predictor of prognosis in DLBCL. To avoid variation, all samples were tested in a single centre and single platform. This study comprised 8 countries (Brazil, Chile, Hungary, India, Philippines, S. Korea, Thailand and Turkey). Using the Kaplan-Meier and log rank test on patients (n=162) and two mortality risk groups (with those above and below the mean representing high and low risk groups) confirmed that the 6-gene predictor score correlates significantly with overall survival (OS, p<0.01) but not with event free survival (EFS, p=0.18). Adding the International Prognostic Index (IPI) shows that the 6-gene predictor score correlates significantly with high IPI scores for OS (p<0.05), whereas those with low IPI scores show a trend not reaching significance (p=0.08). This study defined an effective and economical qRT-PCR strategy and validated the 6-gene score as a predictor of OS in an international setting.
Collapse
Affiliation(s)
- Nilgun Tekin
- Ankara University, Biotechnology Institute, Ankara, Turkey
| | - Nader Omidvar
- Department of Hematology, University of Cardiff School of Medicine, UK
| | - Tim Peter Morris
- Medical Research Council (MRC) Clinical Trials Unit at University College (UCL), London, UK
| | - Paulette Conget
- Facultad de Medicina Clínica Alemana - Universidad del Desarrollo Santiago, Chile
| | - Flavia Bruna
- Facultad de Medicina Clínica Alemana - Universidad del Desarrollo Santiago, Chile
| | - Botond Timar
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Eva Gagyi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ranjan Basak
- Departments of Medical Oncology & Pathology, Tata Memorial Hospital, Mumbai, India
| | - Omkar Naik
- Departments of Medical Oncology & Pathology, Tata Memorial Hospital, Mumbai, India
| | - Chirayu Auewarakul
- Chulabhorn Cancer Centre and Faculty of Medicine Siriraj Hospital, Bangkok, Thailand
| | - Narongrit Sritana
- Chulabhorn Cancer Centre and Faculty of Medicine Siriraj Hospital, Bangkok, Thailand
| | - Debora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, São Paulo/SP, Brazil
| | - Juliano Julio Cerci
- Department of Nuclear Medicine, Quanta - Diagnóstico e Terapia, Curitiba, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, São Paulo/SP, Brazil
| | - Juliana Pereira
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, São Paulo/SP, Brazil
| | - Mark Pierre Dimamay
- Research and Biotechnology Division, St Luke's Medical Centre, Manila, Philippines
| | - Filipinas Natividad
- Research and Biotechnology Division, St Luke's Medical Centre, Manila, Philippines
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Nevin Belder
- Ankara University, Biotechnology Institute, Ankara, Turkey
| | - Isinsu Kuzu
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Diana Paez
- Department of Nuclear Sciences and Application, Division of Human Health, International Atomic Energy Agency, Vienna, Austria
| | - Maurizio Dondi
- Department of Nuclear Sciences and Application, Division of Human Health, International Atomic Energy Agency, Vienna, Austria
| | - Robert Carr
- Department of Haematology, Guy's & St Thomas' Hospital, King's College, London, UK
| | - Hilal Ozdag
- Ankara University, Biotechnology Institute, Ankara, Turkey
| | - Rose Ann Padua
- Institut National de la Sante et de la Recherche Médicale (INSERM) Unité 1131, Université Paris-Diderot, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
23
|
Morgan MM, Johnson BP, Livingston MK, Schuler LA, Alarid ET, Sung KE, Beebe DJ. Personalized in vitro cancer models to predict therapeutic response: Challenges and a framework for improvement. Pharmacol Ther 2016; 165:79-92. [PMID: 27218886 PMCID: PMC5439438 DOI: 10.1016/j.pharmthera.2016.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.
Collapse
Affiliation(s)
- Molly M Morgan
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Brian P Johnson
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Megan K Livingston
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Kyung E Sung
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States.
| | - David J Beebe
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
24
|
Vlasova M, Smirin BV. Personalized Approach in Nanomedicine. ADVANCES IN MEDICAL TECHNOLOGIES AND CLINICAL PRACTICE 2016. [DOI: 10.4018/978-1-5225-0754-3.ch001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This chapter characterizes need for new patient-specific approaches in medicine. The authors here illustrate role of nanomedicine and particularly nanotheranostics, combining diagnostic and therapeutic functions, in the paradigm of personalized disease treatment. This chapter discusses current insights regarding the mechanisms of nano-bio interactions and the origin of adverse effects of nanoformulations. Furthermore, this chapter illustrates possible reasons behind an individual physiological response to a given nanomedicine, such as type and stage of disease, physiological conditions and lifestyle of a patient. Finally, a review of possible approaches for the initial choice of nanoformulation, suitable for a given patient is provided at the end of the chapter.
Collapse
|
25
|
A Balanced Tissue Composition Reveals New Metabolic and Gene Expression Markers in Prostate Cancer. PLoS One 2016; 11:e0153727. [PMID: 27100877 PMCID: PMC4839647 DOI: 10.1371/journal.pone.0153727] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/01/2016] [Indexed: 11/24/2022] Open
Abstract
Molecular analysis of patient tissue samples is essential to characterize the in vivo variability in human cancers which are not accessible in cell-lines or animal models. This applies particularly to studies of tumor metabolism. The challenge is, however, the complex mixture of various tissue types within each sample, such as benign epithelium, stroma and cancer tissue, which can introduce systematic biases when cancers are compared to normal samples. In this study we apply a simple strategy to remove such biases using sample selections where the average content of stroma tissue is balanced between the sample groups. The strategy is applied to a prostate cancer patient cohort where data from MR spectroscopy and gene expression have been collected from and integrated on the exact same tissue samples. We reveal in vivo changes in cancer-relevant metabolic pathways which are otherwise hidden in the data due to tissue confounding. In particular, lowered levels of putrescine are connected to increased expression of SRM, reduced levels of citrate are attributed to upregulation of genes promoting fatty acid synthesis, and increased succinate levels coincide with reduced expression of SUCLA2 and SDHD. In addition, the strategy also highlights important metabolic differences between the stroma, epithelium and prostate cancer. These results show that important in vivo metabolic features of cancer can be revealed from patient data only if the heterogeneous tissue composition is properly accounted for in the analysis.
Collapse
|
26
|
Xie QY, Almudevar A, Whitney-Miller CL, Barry CT, McCall MN. A microRNA biomarker of hepatocellular carcinoma recurrence following liver transplantation accounting for within-patient heterogeneity. BMC Med Genomics 2016; 9:18. [PMID: 27059462 PMCID: PMC4826548 DOI: 10.1186/s12920-016-0179-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/21/2016] [Indexed: 01/06/2023] Open
Abstract
Background Liver cancer, of which hepatocellular carcinoma (HCC) is by far the most common type, is the second most deadly cancer (746,000 deaths in 2012). Currently, the only curative treatment for HCC is surgery to remove the malignancy (resection) or to remove the entire diseased liver followed by transplantation of healthy liver tissue. Given the shortage of healthy livers, it is crucial to provide transplants to patients that have the best chance of long-term survival. Currently, transplantation is determined via the Milan criteria—patients within Milan (single tumor < 5 cm or 2–3 tumors < 3 cm with no extrahepatic spread nor intrahepatic vascular invasion) are typically eligible for transplantation. However, combining microRNA expression profiling with the Milan criteria can improve prediction of recurrence. HCC often presents with multiple distinct tumor foci arising from local spread of a primary tumor or from the oncogenic predisposition of the diseased liver. Substantial genomic heterogeneity between tumor foci within a single patient has been reported; therefore, biomarker development must account for the possibility of highly heterogeneous genomic profiles from the same individual. Methods MicroRNA profiling was performed on 180 HCC tumor samples from 89 patients who underwent liver transplantation at the University of Rochester Medical Center. The primary outcome was recurrence-free survival time, and patients were observed for 3 years post-transplantation. Results MicroRNA expression profiles were used to develop a biomarker that distinguishes HCC patients at greater risk of recurrence post-transplantation. Unsupervised clustering uncovered two distinct subgroups with vast differences in standard transplantation selection criteria and recurrence-free survival times. These subgroups were subsequently used to identify microRNAs strongly associated with HCC recurrence. Our results show that reduced expression of five specific microRNAs is significantly associated with HCC recurrence post-transplantation. Conclusions MicroRNA profiling of distinct tumor foci, coupled with methods that address within-subject tumor heterogeneity, has the potential to significantly improve prediction of HCC recurrence post-transplantation. The development of a clinically applicable HCC biomarker would inform treatment options for patients and contribute to liver transplant selection criteria for practitioners. Electronic supplementary material The online version of this article (doi:10.1186/s12920-016-0179-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Yan Xie
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony Almudevar
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Christopher T Barry
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
27
|
Mukherjee S, Ma Z, Wheeler S, Sathanoori M, Coldren C, Prescott JL, Kozyr N, Bouzyk M, Correll M, Ho H, Chandra PK, Lennon PA. Chromosomal microarray provides enhanced targetable gene aberration detection when paired with next generation sequencing panel in profiling lung and colorectal tumors. Cancer Genet 2016; 209:119-29. [PMID: 26880400 DOI: 10.1016/j.cancergen.2015.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/24/2015] [Accepted: 12/27/2015] [Indexed: 12/12/2022]
Abstract
The development of targeted therapies based on specific genomic alterations has altered the treatment and management of lung and colorectal cancers. Chromosomal microarray (CMA) has allowed identification of copy number variations (CNVs) in lung and colorectal cancers in great detail, and next-generation sequencing (NGS) is used extensively to analyze the genome of cancers for molecular subtyping and use of molecularly guided therapies. The main objective of this study was to evaluate the utility of combining CMA and NGS for a comprehensive genomic assessment of lung and colorectal adenocarcinomas, especially for detecting drug targets. We compared the results from NGS and CMA data from 60 lung and 51 colorectal tumors. From CMA analysis, 33% were amplified, 89% showed gains, 75% showed losses and 41% demonstrated loss of heterozygosity; pathogenic variants were identified in 81% of colon and 67% lung specimens through NGS. KRAS mutations commonly occurred with loss in TP53 and there was significant loss of BRCA1 and NF1 among male patients with lung cancer. For clinically actionable targets, 23% had targetable CNVs when no pathogenic variants were detected by NGS. The data thus indicate that combining the two approaches provides significant benefit in a routine clinical setting not available by NGS alone.
Collapse
Affiliation(s)
| | - Z Ma
- PathGroup, Nashville, TN, USA
| | | | | | | | | | | | | | | | - H Ho
- PathGroup, Nashville, TN, USA
| | | | | |
Collapse
|
28
|
Field MA, Cho V, Andrews TD, Goodnow CC. Reliably Detecting Clinically Important Variants Requires Both Combined Variant Calls and Optimized Filtering Strategies. PLoS One 2015; 10:e0143199. [PMID: 26600436 PMCID: PMC4658170 DOI: 10.1371/journal.pone.0143199] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/02/2015] [Indexed: 12/21/2022] Open
Abstract
A diversity of tools is available for identification of variants from genome sequence data. Given the current complexity of incorporating external software into a genome analysis infrastructure, a tendency exists to rely on the results from a single tool alone. The quality of the output variant calls is highly variable however, depending on factors such as sequence library quality as well as the choice of short-read aligner, variant caller, and variant caller filtering strategy. Here we present a two-part study first using the high quality 'genome in a bottle' reference set to demonstrate the significant impact the choice of aligner, variant caller, and variant caller filtering strategy has on overall variant call quality and further how certain variant callers outperform others with increased sample contamination, an important consideration when analyzing sequenced cancer samples. This analysis confirms previous work showing that combining variant calls of multiple tools results in the best quality resultant variant set, for either specificity or sensitivity, depending on whether the intersection or union, of all variant calls is used respectively. Second, we analyze a melanoma cell line derived from a control lymphocyte sample to determine whether software choices affect the detection of clinically important melanoma risk-factor variants finding that only one of the three such variants is unanimously detected under all conditions. Finally, we describe a cogent strategy for implementing a clinical variant detection pipeline; a strategy that requires careful software selection, variant caller filtering optimizing, and combined variant calls in order to effectively minimize false negative variants. While implementing such features represents an increase in complexity and computation the results offer indisputable improvements in data quality.
Collapse
Affiliation(s)
- Matthew A. Field
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- National Computational Infrastructure, Australian National University, Canberra, ACT, Australia
| | - Vicky Cho
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Australian Phenomics Facility, Australian National University, Canberra, ACT, Australia
| | - T. Daniel Andrews
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- National Computational Infrastructure, Australian National University, Canberra, ACT, Australia
| | - Chris C. Goodnow
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Immunogenomics Group, Immunology Research Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
29
|
Shen Q, Hu J, Jiang N, Hu X, Luo Z, Zhang H. contamDE: differential expression analysis of RNA-seq data for contaminated tumor samples. Bioinformatics 2015; 32:705-12. [DOI: 10.1093/bioinformatics/btv657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 11/03/2015] [Indexed: 11/14/2022] Open
|
30
|
Gupta MK, Bajpai J, Bajpai AK. Optimizing the release process and modelling of in vitro release data of cis-dichlorodiamminoplatinum (II) encapsulated into poly(2-hydroxyethyl methacrylate) nanocarriers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 58:852-62. [PMID: 26478380 DOI: 10.1016/j.msec.2015.09.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/09/2015] [Accepted: 09/10/2015] [Indexed: 12/26/2022]
Abstract
Drug encapsulated nanocarriers are vehicles to transport the drug molecules and release them at the immediate vicinity of the diseased sites. The aim of this study was to design poly (2-hydroxyethyl methacrylate) nanoparticles (PHEMANPs) as a swelling and diffusion controlled drug release system for achieving sustained release of (cis-dichlorodiamminoplatinum II) CDDP. The study undertakes designing and characterization of nanocarriers, optimization of drug encapsulation, and investigating release dynamics of the CDDP drug. PHEMANPs were prepared by suspension polymerization method followed by post loading of the CDDP onto the nanocarriers. The physicochemical and biopharmaceutical properties were evaluated by FTIR, TEM, FESEM, EDX, DLS, surface charge, water intake studies, in vitro cytotoxicity, protein adsorption and percent haemolysis. Chemical stability of the drug was assessed and in vitro release experiments were performed to optimize formulation by UV spectral analysis. The obtained cumulative release data were fitted to zero, first and Korsmeyer-Peppas kinetic models to gain insights into release kinetics and prevailing drug transport mechanisms. The successful encapsulation of CDDP was achieved in different PHEMANP formulations with maximum drug encapsulation efficiency of approx. 60% and the release kinetics was found to follow the Korsmeyer-Peppas model having non-Fickian mechanism. The results indicated that the CDDP can be formulated with a high payload of PHEMANPs which can serve as promising nanomedicine and help in achieving sustained delivery of drug for targeting tumour.
Collapse
Affiliation(s)
- Meher Kanta Gupta
- Bose Memorial Research Laboratory, Department of Chemistry, Govt. Model Science (Autonomous) College, Jabalpur, M.P., 482001, India
| | - Jaya Bajpai
- Bose Memorial Research Laboratory, Department of Chemistry, Govt. Model Science (Autonomous) College, Jabalpur, M.P., 482001, India
| | - Anil Kumar Bajpai
- Bose Memorial Research Laboratory, Department of Chemistry, Govt. Model Science (Autonomous) College, Jabalpur, M.P., 482001, India.
| |
Collapse
|
31
|
Valera E, Shia WW, Bailey RC. Development and validation of an immunosensor for monocyte chemotactic protein 1 using a silicon photonic microring resonator biosensing platform. Clin Biochem 2015; 49:121-6. [PMID: 26365696 DOI: 10.1016/j.clinbiochem.2015.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 09/03/2015] [Accepted: 09/06/2015] [Indexed: 01/20/2023]
Abstract
OBJECTIVES We report the development of an optical immunosensor for the detection of monocyte chemotactic protein 1 (MCP-1) in serum samples. MCP-1 is a cytokine that is an emerging biomarker for several diseases/disorders, including ischemic cardiomyopathy, fibromyalgia, and some cancers. DESIGN AND METHODS The detection of MCP-1 was achieved by performing a sandwich immunoassay on a silicon photonic microring resonator sensor platform. The resonance wavelengths supported by microring sensors are responsive to local changes in the environment accompanying biomarker binding. This technology offers a modularly multiplexable approach to detecting analyte localization in an antibody-antigen complex at the sensor surface. RESULTS The immunosensor allowed the rapid detection of MCP-1 in buffer and spiked human serum samples. An almost 2 order of magnitude linear range was observed, between 84.3 and 1582.1pg/mL and the limits of blank and detection were determined to be 0.3 and 0.5pg/mL, respectively. The platform's ability to analyze MCP-1 concentrations across a clinically-relevant concentration range was demonstrated. CONCLUSIONS A silicon photonic immunosensor technology was applied to the detection of clinically-relevant concentrations of MCP-1. The performance of the sensor was validated through a broad dynamic range and across a number of suggested clinical cut-off values. Importantly, the intrinsic scalability and rapidity of the technology makes it readily amenable to the simultaneous detection of multiplexed biomarker panels, which is particularly needed for the clinical realization of inflammatory diagnostics.
Collapse
Affiliation(s)
- Enrique Valera
- Department of Chemistry, University of Illinois at Urbana - Champaign, 600 South Matthews Avenue, Urbana, IL 61801, United States
| | - Winnie W Shia
- Department of Chemistry, University of Illinois at Urbana - Champaign, 600 South Matthews Avenue, Urbana, IL 61801, United States
| | - Ryan C Bailey
- Department of Chemistry, University of Illinois at Urbana - Champaign, 600 South Matthews Avenue, Urbana, IL 61801, United States.
| |
Collapse
|
32
|
Varn FS, Ung MH, Lou SK, Cheng C. Integrative analysis of survival-associated gene sets in breast cancer. BMC Med Genomics 2015; 8:11. [PMID: 25881247 PMCID: PMC4359519 DOI: 10.1186/s12920-015-0086-0] [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: 10/16/2014] [Accepted: 02/24/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patient gene expression information has recently become a clinical feature used to evaluate breast cancer prognosis. The emergence of prognostic gene sets that take advantage of these data has led to a rich library of information that can be used to characterize the molecular nature of a patient's cancer. Identifying robust gene sets that are consistently predictive of a patient's clinical outcome has become one of the main challenges in the field. METHODS We inputted our previously established BASE algorithm with patient gene expression data and gene sets from MSigDB to develop the gene set activity score (GSAS), a metric that quantitatively assesses a gene set's activity level in a given patient. We utilized this metric, along with patient time-to-event data, to perform survival analyses to identify the gene sets that were significantly correlated with patient survival. We then performed cross-dataset analyses to identify robust prognostic gene sets and to classify patients by metastasis status. Additionally, we created a gene set network based on component gene overlap to explore the relationship between gene sets derived from MSigDB. We developed a novel gene set based on this network's topology and applied the GSAS metric to characterize its role in patient survival. RESULTS Using the GSAS metric, we identified 120 gene sets that were significantly associated with patient survival in all datasets tested. The gene overlap network analysis yielded a novel gene set enriched in genes shared by the robustly predictive gene sets. This gene set was highly correlated to patient survival when used alone. Most interestingly, removal of the genes in this gene set from the gene pool on MSigDB resulted in a large reduction in the number of predictive gene sets, suggesting a prominent role for these genes in breast cancer progression. CONCLUSIONS The GSAS metric provided a useful medium by which we systematically investigated how gene sets from MSigDB relate to breast cancer patient survival. We used this metric to identify predictive gene sets and to construct a novel gene set containing genes heavily involved in cancer progression.
Collapse
Affiliation(s)
- Frederick S Varn
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755, USA.
| | - Matthew H Ung
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755, USA.
| | - Shao Ke Lou
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755, USA.
| | - Chao Cheng
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755, USA. .,Institute for Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03766, USA. .,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03766, USA.
| |
Collapse
|
33
|
Rapid mass spectrometric conversion of tissue biopsy samples into permanent quantitative digital proteome maps. Nat Med 2015; 21:407-13. [PMID: 25730263 PMCID: PMC4390165 DOI: 10.1038/nm.3807] [Citation(s) in RCA: 295] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/20/2015] [Indexed: 02/07/2023]
Abstract
Clinical specimens are each inherently unique, limited and non-renewable. As such, small samples such as tissue biopsies are often completely consumed after a limited number of analyses. Here we present a method that enables fast and reproducible conversion of a small amount of tissue (approximating the quantity obtained by a biopsy) into a single, permanent digital file representing the mass spectrometry-measurable proteome of the sample. The method combines pressure cycling technology (PCT) and SWATH mass spectrometry (MS), and the resulting proteome maps can be analyzed, re-analyzed, compared and mined in silico to detect and quantify specific proteins across multiple samples. We used this method to process and convert 18 biopsy samples from 9 renal cell carcinoma patients into SWATH-MS fragment ion maps. From these proteome maps we detected and quantified more than 2,000 proteins with a high degree of reproducibility across all samples. The identified proteins clearly separated tumorous kidney tissues from healthy tissue, and differentiated distinct histomorphological kidney cancer subtypes.
Collapse
|
34
|
Santacatterina F, Chamorro M, de Arenas CN, Navarro C, Martín MA, Cuezva JM, Sánchez-Aragó M. Quantitative analysis of proteins of metabolism by reverse phase protein microarrays identifies potential biomarkers of rare neuromuscular diseases. J Transl Med 2015; 13:65. [PMID: 25880557 PMCID: PMC4342896 DOI: 10.1186/s12967-015-0424-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/30/2015] [Indexed: 11/16/2022] Open
Abstract
Background Muscle diseases have been associated with changes in the expression of proteins involved in energy metabolism. To this aim we have developed a number of monoclonal antibodies against proteins of energy metabolism. Methods Herein, we have used Reverse Phase Protein Microarrays (RPMA), a high throughput technique, to investigate quantitative changes in protein expression with the aim of identifying potential biomarkers in rare neuromuscular diseases. A cohort of 73 muscle biopsies that included samples from patients diagnosed of Duchenne (DMD), Becker (BMD), symptomatic forms of DMD and BMD in female carriers (Xp21 Carriers), Limb Girdle Muscular Dystrophy Type 2C (LGMD2C), neuronal ceroid lipofuscinosis (NCL), glycogenosis type V (Mc Ardle disease), isolated mitochondrial complex I deficiency, intensive care unit myopathy and control donors were investigated. The nineteen proteins of energy metabolism studied included members of the mitochondrial oxidation of pyruvate, the tricarboxylic acid cycle, β-oxidation of fatty acids, electron transport and oxidative phosphorylation, glycogen metabolism, glycolysis and oxidative stress using highly specific antibodies. Results The results indicate that the phenotype of energy metabolism offers potential biomarkers that could be implemented to refine the understanding of the biological principles of rare diseases and, eventually, the management of these patients. Conclusions We suggest that some biomarkers of energy metabolism could be translated into the clinics to contribute to the improvement of the clinical handling of patients affected by rare diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0424-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Margarita Chamorro
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Carmen Navarro
- Instituto de Investigación Biomédico de Vigo (IBIV), Hospital Universitario de Vigo, Meixoeiro, 36200, Vigo, Spain.
| | - Miguel A Martín
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain. .,Laboratorio de Enfermedades Mitocondriales y Neuromusculares, Hospital Universitario 12 de Octubre, 28041, Madrid, Spain.
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| |
Collapse
|
35
|
Augsburger JJ, Corrêa ZM, Augsburger BD. Frequency and implications of discordant gene expression profile class in posterior uveal melanomas sampled by fine needle aspiration biopsy. Am J Ophthalmol 2015; 159:248-56. [PMID: 25448994 DOI: 10.1016/j.ajo.2014.10.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE To determine the frequency of discordant gene expression profile (GEP) classification of posterior uveal melanomas sampled at 2 tumor sites by fine-needle aspiration biopsy (FNAB). DESIGN Prospective single-institution longitudinal study performed in conjunction with a multicenter validation study of the prognostic value of GEP class of posterior uveal melanoma cells for metastasis and metastatic death. METHODS FNAB aspirates of 80 clinically diagnosed primary choroidal and ciliochoroidal melanomas were obtained from 2 tumor sites prior to or at the time of initial ocular tumor treatment and submitted for independent GEP testing and classification. Frequency of discordant GEP classification of these specimens was determined. RESULTS Using the support vector machine learning algorithm favored by the developer of the GEP test employed in this study, 9 of the 80 cases (11.3% [95% confidence interval: 9.0%-13.6%]) were clearly discordant. If cases with a failed classification at 1 site or a low confidence class assignment by the support vector machine algorithm at 1 or both sites are also regarded as discordant, then this frequency rises to 13 of the 80 cases (16.3% [95% confidence interval: 13.0%-19.6%]). CONCLUSION Sampling of a clinically diagnosed posterior uveal melanoma at a single site for prognostic GEP testing is associated with a substantial probability of misclassification. Two-site sampling of such tumors with independent GEP testing of each specimen may be advisable to lessen the probability of underestimating an individual patient's prognostic risk of metastasis and metastatic death.
Collapse
|
36
|
Proteomic expressional profiling of a paraffin-embedded tissue by multiplex tissue immunoblotting. Methods Mol Biol 2015; 1312:175-84. [PMID: 26044002 DOI: 10.1007/978-1-4939-2694-7_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the functional proteome era, the proteomic profiling of clinicopathologic annotated tissues is an essential step for mining and evaluations of candidate biomarkers for disease. Previously, application of routine proteomic methodologies to clinical tissue specimens has provided unsatisfactory results. Multiplex tissue immunoblotting is a method of transferring proteins from a formalin-fixed, paraffin-embedded tissue section to a stack of membranes which can be applied to a conventional immunoblotting method. A single tissue section can be transferred to up to ten membranes, each of which is probed with antibodies and detected with fluorescent tags. By this approach, total protein and target signals can be simultaneously determined on each membrane; hence each antibody is internally normalized. Phosphorylation specific antibodies as well as antibodies that do not readily work well with paraffin-embedded tissue are applicable to the membranes, expanding the menu of antibodies that can be utilized with formalin-fixed tissue. This novel platform can provide quantitative detection retaining histomorphologic detail in clinical samples and has great potential to facilitate discovery and development of new diagnostic assays and therapeutic agents.
Collapse
|
37
|
Crosetto N, Bienko M, van Oudenaarden A. Spatially resolved transcriptomics and beyond. Nat Rev Genet 2014; 16:57-66. [DOI: 10.1038/nrg3832] [Citation(s) in RCA: 323] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Khaleel SS, Andrews EH, Ung M, DiRenzo J, Cheng C. E2F4 regulatory program predicts patient survival prognosis in breast cancer. Breast Cancer Res 2014; 16:486. [PMID: 25440089 PMCID: PMC4303196 DOI: 10.1186/s13058-014-0486-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 11/18/2014] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Genetic and molecular signatures have been incorporated into cancer prognosis prediction and treatment decisions with good success over the past decade. Clinically, these signatures are usually used in early-stage cancers to evaluate whether they require adjuvant therapy following surgical resection. A molecular signature that is prognostic across more clinical contexts would be a useful addition to current signatures. METHODS We defined a signature for the ubiquitous tissue factor, E2F4, based on its shared target genes in multiple tissues. These target genes were identified by chromatin immunoprecipitation sequencing (ChIP-seq) experiments using a probabilistic method. We then computationally calculated the regulatory activity score (RAS) of E2F4 in cancer tissues, and examined how E2F4 RAS correlates with patient survival. RESULTS Genes in our E2F4 signature were 21-fold more likely to be correlated with breast cancer patient survival time compared to randomly selected genes. Using eight independent breast cancer datasets containing over 1,900 unique samples, we stratified patients into low and high E2F4 RAS groups. E2F4 activity stratification was highly predictive of patient outcome, and our results remained robust even when controlling for many factors including patient age, tumor size, grade, estrogen receptor (ER) status, lymph node (LN) status, whether the patient received adjuvant therapy, and the patient's other prognostic indices such as Adjuvant! and the Nottingham Prognostic Index scores. Furthermore, the fractions of samples with positive E2F4 RAS vary in different intrinsic breast cancer subtypes, consistent with the different survival profiles of these subtypes. CONCLUSIONS We defined a prognostic signature, the E2F4 regulatory activity score, and showed it to be significantly predictive of patient outcome in breast cancer regardless of treatment status and the states of many other clinicopathological variables. It can be used in conjunction with other breast cancer classification methods such as Oncotype DX to improve clinical outcome prediction.
Collapse
Affiliation(s)
- Sari S Khaleel
- Department of Genetics, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
| | - Erik H Andrews
- Department of Genetics, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
| | - Matthew Ung
- Department of Genetics, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
| | - James DiRenzo
- Department of Pharmacology & Toxicology, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
| | - Chao Cheng
- Department of Genetics, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
- Institute for Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, 03766, USA.
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, 03766, USA.
| |
Collapse
|
39
|
Prediction of individual response to anticancer therapy: historical and future perspectives. Cell Mol Life Sci 2014; 72:729-57. [PMID: 25387856 PMCID: PMC4309902 DOI: 10.1007/s00018-014-1772-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 10/23/2014] [Accepted: 10/27/2014] [Indexed: 02/06/2023]
Abstract
Since the introduction of chemotherapy for cancer treatment in the early 20th century considerable efforts have been made to maximize drug efficiency and at the same time minimize side effects. As there is a great interpatient variability in response to chemotherapy, the development of predictive biomarkers is an ambitious aim for the rapidly growing research area of personalized molecular medicine. The individual prediction of response will improve treatment and thus increase survival and life quality of patients. In the past, cell cultures were used as in vitro models to predict in vivo response to chemotherapy. Several in vitro chemosensitivity assays served as tools to measure miscellaneous endpoints such as DNA damage, apoptosis and cytotoxicity or growth inhibition. Twenty years ago, the development of high-throughput technologies, e.g. cDNA microarrays enabled a more detailed analysis of drug responses. Thousands of genes were screened and expression levels were correlated to drug responses. In addition, mutation analysis became more and more important for the prediction of therapeutic success. Today, as research enters the area of -omics technologies, identification of signaling pathways is a tool to understand molecular mechanism underlying drug resistance. Combining new tissue models, e.g. 3D organoid cultures with modern technologies for biomarker discovery will offer new opportunities to identify new drug targets and in parallel predict individual responses to anticancer therapy. In this review, we present different currently used chemosensitivity assays including 2D and 3D cell culture models and several -omics approaches for the discovery of predictive biomarkers. Furthermore, we discuss the potential of these assays and biomarkers to predict the clinical outcome of individual patients and future perspectives.
Collapse
|
40
|
Targeted nanoparticles for simultaneous delivery of chemotherapeutic and hyperthermia agents – An in vitro study. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 136:81-90. [DOI: 10.1016/j.jphotobiol.2014.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/10/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022]
|
41
|
Su X, Dong C, Zhang J, Su L, Wang X, Cui H, Chen Z. Combination therapy of anti-cancer bioactive peptide with Cisplatin decreases chemotherapy dosing and toxicity to improve the quality of life in xenograft nude mice bearing human gastric cancer. Cell Biosci 2014; 4:7. [PMID: 24507386 PMCID: PMC3930002 DOI: 10.1186/2045-3701-4-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/13/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A great challenge of cancer chemotherapy is to eliminate cancer cells and concurrently maintain the quality of life (QOL) for cancer patients. Previously, we identified a novel anti-cancer bioactive peptide (ACBP), a peptide induced in goat spleen or liver following immunization with human gastric cancer protein extract. ACBP alone exhibited anti-tumor activity without measurable side effects. Thus, we hypothesize that ACBP and combined chemotherapy could improve the efficacy of treatment and lead to a better QOL. RESULTS In this study, ACBP was isolated and purified from immunized goat liver, and designated as ACBP-L. The anti-tumor activity was investigated in a previously untested human gastric cancer MGC-803 cell line and tumor model. ACBP-L inhibited cell proliferation in vitro in a dose and time dependent manner, titrated by MTT assay. The effect of ACBP-L on cell morphology was observed through light and scanning electron microscopy. In vivo ACBP-L alone significantly inhibited MGC-803 tumor growth in a xenograft nude mouse model without measurable side effects. Treatment with the full dosage of Cisplatin alone (5 mg/kg every 5 days) strongly suppressed tumor growth. However, the QOL in these mice had been significantly affected when measured by food intakes and body weight. The combinatory regiment of ACBP-L with a fewer doses of Cisplatin (5 mg/kg every 10 days) resulted in a similar anti-tumor activity with improved QOL. 18F-FDG PET/CT scan was used to examine the biological activity in tumors of live animals and indicated the consistent treatment effects. The tumor tissues were harvested after treatment, and ACBP-L and Cisplatin treatment suppressed Bcl-2, and induced Bax, Caspase 3, and Caspase 8 molecules as detected by RT-PCR and immunohistochemistry. The combinatory regiment induced stronger Bax and Caspase 8 protein expression. CONCLUSION Our current finding in this gastric cancer xenograft animal model demonstrated that ACBP-L could lower Cisplatin dose to achieve a similar anti-tumor efficacy as the higher dose of Cisplatin alone, through enhanced modulation of apoptotic molecules. This newly developed combination regiment improved QOL in tumor bearing hosts, which could lead to clinical investigation for the new strategy of combination therapy.
Collapse
Affiliation(s)
- Xiulan Su
- Clinical Medicine Research Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Chao Dong
- Clinical Medicine Research Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Jialing Zhang
- Clinical Medicine Research Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Liya Su
- Clinical Medicine Research Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Xuemei Wang
- PET-CT Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010051, China
| | - Hongwei Cui
- Clinical Medicine Research Center of The Affiliated Hospital, Inner Mongolia Medical University, No 1 Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892-1419, USA
| |
Collapse
|
42
|
Kang Y. Functional genomic analysis of cancer metastasis: biologic insights and clinical implications. Expert Rev Mol Diagn 2014; 5:385-95. [PMID: 15934815 DOI: 10.1586/14737159.5.3.385] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Metastasis, the spread of cancer from primary tumors to distant vital organs, has devastating consequences. Lack of effective tools to study this complex problem has hindered the development of accurate prognostic methods and effective treatments for metastatic cancer. In the postgenomic era, the application of genomic profiling methods to the analysis of clinical metastasis samples and animal metastasis models has revolutionized the field of metastasis research. This article reviews recent breakthroughs in the functional genomic analysis of metastasis. In addition, its impacts on our understanding of the molecular basis of metastasis and on clinical practice are discussed.
Collapse
Affiliation(s)
- Yibin Kang
- Princeton University Department of Molecular Biology, Princeton, NJ 08544, USA.
| |
Collapse
|
43
|
Zhang Q, Reed EF. Array-based methods for diagnosis and prevention of transplant rejection. Expert Rev Mol Diagn 2014; 6:165-78. [PMID: 16512777 DOI: 10.1586/14737159.6.2.165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
DNA microarray is a microhybridization-based assay that is used to simultaneously study the expression of thousands of genes, thus providing a global view of gene expression in a tissue sample. This powerful technique has been adopted by many biomedical disciplines and will likely have a profound impact on the diagnosis, treatment and prognosis of human diseases. This review article presents an overview of the application of microarray technology to the field of solid-organ transplantation.
Collapse
Affiliation(s)
- Qiuheng Zhang
- Immunogenetics Center, Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
44
|
Ewis AA, Zhelev Z, Bakalova R, Fukuoka S, Shinohara Y, Ishikawa M, Baba Y. A history of microarrays in biomedicine. Expert Rev Mol Diagn 2014; 5:315-28. [PMID: 15934810 DOI: 10.1586/14737159.5.3.315] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The fundamental strategy of the current postgenomic era or the era of functional genomics is to expand the scale of biologic research from studying single genes or proteins to studying all genes or proteins simultaneously using a systematic approach. As recently developed methods for obtaining genome-wide mRNA expression data, oligonucleotide and DNA microarrays are particularly powerful in the context of knowing the entire genome sequence and can provide a global view of changes in gene expression patterns in response to physiologic alterations or manipulation of transcriptional regulators. In biomedical research, such an approach will ultimately determine biologic behavior of both normal and diseased tissues, which may provide insights into disease mechanisms and identify novel markers and candidates for diagnostic, prognostic and therapeutic intervention. However, microarray technology is still in a continuous state of evolution and development, and it may take time to implement microarrays as a routine medical device. Many limitations exist and many challenges remain to be achieved to help inclusion of microarrays in clinical medicine. In this review, a brief history of microarrays in biomedical research is provided, including experimental overview, limitations, challenges and future developments.
Collapse
Affiliation(s)
- Ashraf A Ewis
- Single-Molecule Bioanalysis Laboratory, National Institute of Advanced Industrial Science & Technology (AIST), Hayashi-cho 2217-14, Takamatsu City, Kagawa Prefecture, 761-0395 Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Jeon J, Nim S, Teyra J, Datti A, Wrana JL, Sidhu SS, Moffat J, Kim PM. A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening. Genome Med 2014; 6:57. [PMID: 25165489 PMCID: PMC4143549 DOI: 10.1186/s13073-014-0057-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/18/2014] [Indexed: 12/14/2022] Open
Abstract
We present an integrated approach that predicts and validates novel anti-cancer drug targets. We first built a classifier that integrates a variety of genomic and systematic datasets to prioritize drug targets specific for breast, pancreatic and ovarian cancer. We then devised strategies to inhibit these anti-cancer drug targets and selected a set of targets that are amenable to inhibition by small molecules, antibodies and synthetic peptides. We validated the predicted drug targets by showing strong anti-proliferative effects of both synthetic peptide and small molecule inhibitors against our predicted targets.
Collapse
Affiliation(s)
- Jouhyun Jeon
| | - Satra Nim
| | - Joan Teyra
| | - Alessandro Datti
| | - Jeffrey L Wrana
| | - Sachdev S Sidhu
| | - Jason Moffat
| | - Philip M Kim
| |
Collapse
|
46
|
Nikolaishvilli-Feinberg N, Cohen SM, Midkiff B, Zhou Y, Olorvida M, Ibrahim JG, Omolo B, Shields JM, Thomas NE, Groben PA, Kaufmann WK, Miller CR. Development of DNA damage response signaling biomarkers using automated, quantitative image analysis. J Histochem Cytochem 2013; 62:185-96. [PMID: 24309508 DOI: 10.1369/0022155413516469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The DNA damage response (DDR) coordinates DNA repair with cell cycle checkpoints to ameliorate or mitigate the pathological effects of DNA damage. Automated quantitative analysis (AQUA) and Tissue Studio are commercial technologies that use digitized immunofluorescence microscopy images to quantify antigen expression in defined tissue compartments. Because DDR is commonly activated in cancer and may reflect genetic instability within the lesion, a method to quantify DDR in cancer offers potential diagnostic and/or prognostic value. In this study, both AQUA and Tissue Studio algorithms were used to quantify the DDR in radiation-damaged skin fibroblasts, melanoma cell lines, moles, and primary and metastatic melanomas. Digital image analysis results for three markers of DDR (γH2AX, P-ATM, P-Chk2) correlated with immunoblot data for irradiated fibroblasts, whereas only γH2AX and P-Chk2 correlated with immunoblot data in melanoma cell lines. Melanoma cell lines displayed substantial variation in γH2AX and P-Chk2 expression, and P-Chk2 expression was significantly correlated with radioresistance. Moles, primary melanomas, and melanoma metastases in brain, lung and liver displayed substantial variation in γH2AX expression, similar to that observed in melanoma cell lines. Automated digital analysis of immunofluorescent images stained for DDR biomarkers may be useful for predicting tumor response to radiation and chemotherapy.
Collapse
Affiliation(s)
- Nana Nikolaishvilli-Feinberg
- Translational Pathology Laboratory (NNF, SMC, BM, MO, CRM), University of North Carolina School of Medicine, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kim TM, Lee SH, Chung YJ. Clinical applications of next-generation sequencing in colorectal cancers. World J Gastroenterol 2013; 19:6784-6793. [PMID: 24187453 PMCID: PMC3812477 DOI: 10.3748/wjg.v19.i40.6784] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/22/2013] [Accepted: 08/20/2013] [Indexed: 02/06/2023] Open
Abstract
Like other solid tumors, colorectal cancer (CRC) is a genomic disorder in which various types of genomic alterations, such as point mutations, genomic rearrangements, gene fusions, or chromosomal copy number alterations, can contribute to the initiation and progression of the disease. The advent of a new DNA sequencing technology known as next-generation sequencing (NGS) has revolutionized the speed and throughput of cataloguing such cancer-related genomic alterations. Now the challenge is how to exploit this advanced technology to better understand the underlying molecular mechanism of colorectal carcinogenesis and to identify clinically relevant genetic biomarkers for diagnosis and personalized therapeutics. In this review, we will introduce NGS-based cancer genomics studies focusing on those of CRC, including a recent large-scale report from the Cancer Genome Atlas. We will mainly discuss how NGS-based exome-, whole genome- and methylome-sequencing have extended our understanding of colorectal carcinogenesis. We will also introduce the unique genomic features of CRC discovered by NGS technologies, such as the relationship with bacterial pathogens and the massive genomic rearrangements of chromothripsis. Finally, we will discuss the necessary steps prior to development of a clinical application of NGS-related findings for the advanced management of patients with CRC.
Collapse
|
48
|
Quantum dot imaging platform for single-cell molecular profiling. Nat Commun 2013; 4:1619. [PMID: 23511483 PMCID: PMC3615486 DOI: 10.1038/ncomms2635] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/21/2013] [Indexed: 12/18/2022] Open
Abstract
Study of normal cell physiology and disease pathogenesis heavily relies on untangling the complexity of intracellular molecular mechanisms and pathways. To achieve this goal, comprehensive molecular profiling of individual cells within the context of microenvironment is required. Here we report the development of a multicolour multicycle in situ imaging technology capable of creating detailed quantitative molecular profiles for individual cells at the resolution of optical imaging. A library of stoichiometric fluorescent probes is prepared by linking target-specific antibodies to a universal quantum dot-based platform via protein A in a quick and simple procedure. Surprisingly, despite the potential for multivalent binding between protein A and antibody and the intermediate affinity of this non-covalent bond, fully assembled probes do not aggregate or exchange antibodies, facilitating highly multiplexed parallel staining. This single-cell molecular profiling technology is expected to open new opportunities in systems biology, gene expression studies, signalling pathway analysis and molecular diagnostics. Multiplexed labelling of individual cells allows the direct observation of intracellular molecular composition, but is difficult to achieve with existing techniques. Here, self-assembled fluorescent nanoparticle probes and multicolour multicycle staining are used for the simultaneous evaluation of multiple biomolecules at subcellular resolution.
Collapse
|
49
|
Abstract
Here we present a detailed protocol for molecular profiling of individual cultured mammalian cells using multicolor multicycle immunofluorescence with quantum dot probes. It includes instructions for cell culture growth and processing (2 h + 48-72 h for cell growth), preparation and characterization of universal quantum dot probes (4.5 h + overnight incubation), cyclic cell staining (∼4.5 h per cycle) and image analysis (varies by application). The use of quantum dot fluorescent probes enables highly multiplexed, robust quantitative molecular imaging with a conventional fluorescence microscopy setup, whereas the probe preparation methodology, using a self-assembly between protein A-decorated universal quantum dots and intact primary antibodies, offers a fast, simple and purification-free route for an on-demand preparation of antibody-functionalized quantum dot libraries. As a result, this protocol can be used by biomedical researchers for a variety of cell staining applications, and, with further optimization, for staining of other biological specimens (e.g., clinical tissue sections).
Collapse
|
50
|
|