151
|
Pantel K, Speicher MR. The biology of circulating tumor cells. Oncogene 2016; 35:1216-24. [PMID: 26050619 DOI: 10.1038/onc.2015.192] [Citation(s) in RCA: 370] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 12/15/2022]
Abstract
Metastasis is a biologically complex process consisting of numerous stochastic events which may tremendously differ across various cancer types. Circulating tumor cells (CTCs) are cells that are shed from primary tumors and metastatic deposits into the blood stream. CTCs bear a tremendous potential to improve our understanding of steps involved in the metastatic cascade, starting from intravasation of tumor cells into the circulation until the formation of clinically detectable metastasis. These efforts were propelled by novel high-resolution approaches to dissect the genomes and transcriptomes of CTCs. Furthermore, capturing of viable CTCs has paved the way for innovative culturing technologies to study fundamental characteristics of CTCs such as invasiveness, their kinetics and responses to selection barriers, such as given therapies. Hence the study of CTCs is not only instrumental as a basic research tool, but also allows the serial monitoring of tumor genotypes and may therefore provide predictive and prognostic biomarkers for clinicians. Here, we review how CTCs have contributed to significant insights into the metastatic process and how they may be utilized in clinical practice.
Collapse
Affiliation(s)
- K Pantel
- Institute of Tumor Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - M R Speicher
- Institute of Human Genetics, Medical University of Graz, Graz, Austria
| |
Collapse
|
152
|
Dadiani M, Bossel Ben-Moshe N, Paluch-Shimon S, Perry G, Balint N, Marin I, Pavlovski A, Morzaev D, Kahana-Edwin S, Yosepovich A, Gal-Yam EN, Berger R, Barshack I, Domany E, Kaufman B. Tumor Evolution Inferred by Patterns of microRNA Expression through the Course of Disease, Therapy, and Recurrence in Breast Cancer. Clin Cancer Res 2016; 22:3651-62. [PMID: 26957561 DOI: 10.1158/1078-0432.ccr-15-2313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/21/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Molecular evolution of tumors during progression, therapy, and metastasis is a major clinical challenge and the main reason for resistance to therapy. We hypothesized that microRNAs (miRNAs) that exhibit similar variation of expression through the course of disease in several patients have a significant function in the tumorigenic process. EXPERIMENTAL DESIGN Exploration of evolving disease by profiling 800 miRNA expression from serial samples of individual breast cancer patients at several time points: pretreatment, posttreatment, lymph nodes, and recurrence sites when available (58 unique samples from 19 patients). Using a dynamic approach for analysis, we identified expression modulation patterns and classified varying miRNAs into one of the eight possible temporal expression patterns. RESULTS The various patterns were found to be associated with different tumorigenic pathways. The dominant pattern identified an miRNA set that significantly differentiated between disease stages, and its pattern in each patient was also associated with response to therapy. These miRNAs were related to tumor proliferation and to the cell-cycle pathway, and their mRNA targets showed anticorrelated expression. Interestingly, the level of these miRNAs was lowest in matched recurrent samples from distant metastasis, indicating a gradual increase in proliferative potential through the course of disease. Finally, the average expression level of these miRNAs in the pretreatment biopsy was significantly different comparing patients experiencing recurrence to recurrence-free patients. CONCLUSIONS Serial tumor sampling combined with analysis of temporal expression patterns enabled to pinpoint significant signatures characterizing breast cancer progression, associated with response to therapy and with risk of recurrence. Clin Cancer Res; 22(14); 3651-62. ©2016 AACR.
Collapse
Affiliation(s)
- Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel.
| | - Noa Bossel Ben-Moshe
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | | | - Gili Perry
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Nora Balint
- Pathology Institute, Sheba Medical Center, Tel-Hashomer, Israel
| | - Irina Marin
- Pathology Institute, Sheba Medical Center, Tel-Hashomer, Israel
| | - Anya Pavlovski
- Pathology Institute, Sheba Medical Center, Tel-Hashomer, Israel
| | - Dana Morzaev
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Ady Yosepovich
- Pathology Institute, Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Raanan Berger
- Oncology Institute, Sheba Medical Center, Tel-Hashomer, Israel
| | - Iris Barshack
- Pathology Institute, Sheba Medical Center, Tel-Hashomer, Israel. Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Domany
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Bella Kaufman
- Oncology Institute, Sheba Medical Center, Tel-Hashomer, Israel. Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
153
|
Williams MJ, Werner B, Barnes CP, Graham TA, Sottoriva A. Identification of neutral tumor evolution across cancer types. Nat Genet 2016; 48:238-244. [PMID: 26780609 PMCID: PMC4934603 DOI: 10.1038/ng.3489] [Citation(s) in RCA: 419] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
Despite extraordinary efforts to profile cancer genomes, interpreting the vast amount of genomic data in the light of cancer evolution remains challenging. Here we demonstrate that neutral tumor evolution results in a power-law distribution of the mutant allele frequencies reported by next-generation sequencing of tumor bulk samples. We find that the neutral power law fits with high precision 323 of 904 cancers from 14 types and from different cohorts. In malignancies identified as evolving neutrally, all clonal selection seemingly occurred before the onset of cancer growth and not in later-arising subclones, resulting in numerous passenger mutations that are responsible for intratumoral heterogeneity. Reanalyzing cancer sequencing data within the neutral framework allowed the measurement, in each patient, of both the in vivo mutation rate and the order and timing of mutations. This result provides a new way to interpret existing cancer genomic data and to discriminate between functional and non-functional intratumoral heterogeneity.
Collapse
Affiliation(s)
- Marc J Williams
- Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology (CoMPLEX), University College London, London, WC1E 6BT, UK
| | - Benjamin Werner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
- Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| |
Collapse
|
154
|
Andrade SS, Gouvea IE, Silva MCC, Castro ED, de Paula CAA, Okamoto D, Oliveira L, Peres GB, Ottaiano T, Facina G, Nazário ACP, Campos AHJFM, Paredes-Gamero EJ, Juliano M, da Silva IDCG, Oliva MLV, Girão MJBC. Cathepsin K induces platelet dysfunction and affects cell signaling in breast cancer - molecularly distinct behavior of cathepsin K in breast cancer. BMC Cancer 2016; 16:173. [PMID: 26931461 PMCID: PMC4774035 DOI: 10.1186/s12885-016-2203-7] [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: 09/30/2015] [Accepted: 02/17/2016] [Indexed: 11/12/2022] Open
Abstract
Background Breast cancer comprises clinically and molecularly distinct tumor subgroups that differ in cell histology and biology and show divergent clinical phenotypes that impede phase III trials, such as those utilizing cathepsin K inhibitors. Here we correlate the epithelial-mesenchymal-like transition breast cancer cells and cathepsin K secretion with activation and aggregation of platelets. Cathepsin K is up-regulated in cancer cells that proteolyze extracellular matrix and contributes to invasiveness. Although proteolytically activated receptors (PARs) are activated by proteases, the direct interaction of cysteine cathepsins with PARs is poorly understood. In human platelets, PAR-1 and −4 are highly expressed, but PAR-3 shows low expression and unclear functions. Methods Platelet aggregation was monitored by measuring changes in turbidity. Platelets were immunoblotted with anti-phospho and total p38, Src-Tyr-416, FAK-Tyr-397, and TGFβ monoclonal antibody. Activation was measured in a flow cytometer and calcium mobilization in a confocal microscope. Mammary epithelial cells were prepared from the primary breast cancer samples of 15 women with Luminal-B subtype to produce primary cells. Results We demonstrate that platelets are aggregated by cathepsin K in a dose-dependent manner, but not by other cysteine cathepsins. PARs-3 and −4 were confirmed as the cathepsin K target by immunodetection and specific antagonists using a fibroblast cell line derived from PARs deficient mice. Moreover, through co-culture experiments, we show that platelets activated by cathepsin K mediated the up-regulation of SHH, PTHrP, OPN, and TGFβ in epithelial-mesenchymal-like cells from patients with Luminal B breast cancer. Conclusions Cathepsin K induces platelet dysfunction and affects signaling in breast cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2203-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheila Siqueira Andrade
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil. .,Charitable Association of Blood Collection - COLSAN, São Paulo, SP, 04080-006, Brazil. .,Department of Gynecology, Cellular Gynecology Laboratory, Universidade Federal de São Paulo, Rua Napoleão de Barros, 608, CEP 04024-002, São Paulo, Brazil.
| | - Iuri Estrada Gouvea
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | | | - Eloísa Dognani Castro
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Cláudia A A de Paula
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Debora Okamoto
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Lilian Oliveira
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Giovani Bravin Peres
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Tatiana Ottaiano
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Gil Facina
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | | | - Antonio Hugo J F M Campos
- Department of Pathology, AC Camargo Hospital Biobank, A C Camargo Cancer Center - Antonio Prudente Foundation, São Paulo, SP, 01509-010, Brazil.
| | | | - Maria Juliano
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Ismael D C G da Silva
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Maria Luiza V Oliva
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Manoel J B C Girão
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil. .,Charitable Association of Blood Collection - COLSAN, São Paulo, SP, 04080-006, Brazil.
| |
Collapse
|
155
|
Beerenwinkel N, Greenman CD, Lagergren J. Computational Cancer Biology: An Evolutionary Perspective. PLoS Comput Biol 2016; 12:e1004717. [PMID: 26845763 PMCID: PMC4742235 DOI: 10.1371/journal.pcbi.1004717] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
- * E-mail: (NB); (CDG); (JL)
| | - Chris D. Greenman
- School of Computing Sciences, University of East Anglia, Norwich, United Kingdom
- * E-mail: (NB); (CDG); (JL)
| | - Jens Lagergren
- Science for Life Laboratory, School of Computer Science and Communication, Swedish E-Science Research Center, KTH Royal Institute of Technology, Solna, Sweden
- * E-mail: (NB); (CDG); (JL)
| |
Collapse
|
156
|
Intratumor molecular heterogeneity in pleomorphic adenoma of the salivary glands. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 121:158-63. [DOI: 10.1016/j.oooo.2015.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 02/06/2023]
|
157
|
Imaging tumour cell heterogeneity following cell transplantation into optically clear immune-deficient zebrafish. Nat Commun 2016; 7:10358. [PMID: 26790525 PMCID: PMC4735845 DOI: 10.1038/ncomms10358] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 12/02/2015] [Indexed: 02/07/2023] Open
Abstract
Cancers contain a wide diversity of cell types that are defined by differentiation states, genetic mutations and altered epigenetic programmes that impart functional diversity to individual cells. Elevated tumour cell heterogeneity is linked with progression, therapy resistance and relapse. Yet, imaging of tumour cell heterogeneity and the hallmarks of cancer has been a technical and biological challenge. Here we develop optically clear immune-compromised rag2E450fs(casper) zebrafish for optimized cell transplantation and direct visualization of fluorescently labelled cancer cells at single-cell resolution. Tumour engraftment permits dynamic imaging of neovascularization, niche partitioning of tumour-propagating cells in embryonal rhabdomyosarcoma, emergence of clonal dominance in T-cell acute lymphoblastic leukaemia and tumour evolution resulting in elevated growth and metastasis in BRAFV600E-driven melanoma. Cell transplantation approaches using optically clear immune-compromised zebrafish provide unique opportunities to uncover biology underlying cancer and to dynamically visualize cancer processes at single-cell resolution in vivo. Direct visualisation of heterogeneous cell populations in live animals has been challenging. Here, the authors optimize cell transplantation into optically clear immune-deficient zebrafish, and use intravital imaging to track and to assess functional diversity of individual cancer cells in vivo.
Collapse
|
158
|
Abstract
The field of pathology is rapidly transforming from a semiquantitative and empirical science toward a big data discipline. Large data sets from across multiple omics fields may now be extracted from a patient's tissue sample. Tissue is, however, complex, heterogeneous, and prone to artifact. A reductionist view of tissue and disease progression, which does not take this complexity into account, may lead to single biomarkers failing in clinical trials. The integration of standardized multi-omics big data and the retention of valuable information on spatial heterogeneity are imperative to model complex disease mechanisms. Mathematical modeling through systems pathology approaches is the ideal medium to distill the significant information from these large, multi-parametric, and hierarchical data sets. Systems pathology may also predict the dynamical response of disease progression or response to therapy regimens from a static tissue sample. Next-generation pathology will incorporate big data with systems medicine in order to personalize clinical practice for both prognostic and predictive patient care.
Collapse
Affiliation(s)
- Peter D Caie
- Quantitative and systems pathology, University of St Andrews, North Haugh, Fife, St Andrews, KY16 9TF, UK
| | - David J Harrison
- Quantitative and systems pathology, University of St Andrews, North Haugh, Fife, St Andrews, KY16 9TF, UK.
| |
Collapse
|
159
|
Rida PCG, Cantuaria G, Reid MD, Kucuk O, Aneja R. How to be good at being bad: centrosome amplification and mitotic propensity drive intratumoral heterogeneity. Cancer Metastasis Rev 2015; 34:703-13. [PMID: 26358854 PMCID: PMC4778553 DOI: 10.1007/s10555-015-9590-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cancer is truly an iconic disease--a tour de force whose multiple formidable strengths can be attributed to the bewildering heterogeneity that a tumor can manifest both spatially and temporally. A Darwinian evolutionary process is believed to undergird, at least in part, the generation of this heterogeneity that contributes to poor clinical outcomes. Risk assessment in clinical oncology is currently based on a small number of clinicopathologic factors (like stage, histological grade, receptor status, and serum tumor markers) and offers limited accuracy in predicting disease course as evidenced by the prognostic heterogeneity that persists in risk segments produced by present-day models. We posit that this insufficiency stems from the exclusion of key risk contributors from such models, especially the omission of certain factors implicated in generating intratumoral heterogeneity. The extent of centrosome amplification and the mitotic propensity inherent in a tumor are two such vital factors whose contributions to poor prognosis are presently overlooked in risk prognostication. Supernumerary centrosomes occur widely in tumors and are potent drivers of chromosomal instability that fosters intratumoral heterogeneity. The mitotic propensity of a proliferating population of tumor cells reflects the cell cycling kinetics of that population. Since frequent passage through improperly regulated mitotic divisions accelerates production of diverse genotypes, the mitotic propensity inherent in a tumor serves as a powerful beacon of risk. In this review, we highlight how centrosome amplification and error-prone mitoses contribute to poor clinical outcomes and urge the need to develop these cancer-specific traits as much-needed clinically-facile prognostic biomarkers with immense potential value for individualized cancer treatment in the clinic.
Collapse
Affiliation(s)
- Padmashree C G Rida
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Novazoi Theranostics Inc., Plano, TX, 75025, USA
| | - Guilherme Cantuaria
- Department of Gynecologic Oncology, Northside Hospital Cancer Institute, Atlanta, GA, 30342, USA
| | - Michelle D Reid
- Deparment of Pathology, Emory Univ Hospital, Atlanta, GA, 30033, USA
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
160
|
Andor N, Graham TA, Jansen M, Xia LC, Aktipis CA, Petritsch C, Ji HP, Maley CC. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med 2015; 22:105-13. [PMID: 26618723 DOI: 10.1038/nm.3984] [Citation(s) in RCA: 577] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/07/2015] [Indexed: 02/07/2023]
Abstract
Intratumor heterogeneity (ITH) drives neoplastic progression and therapeutic resistance. We used the bioinformatics tools 'expanding ploidy and allele frequency on nested subpopulations' (EXPANDS) and PyClone to detect clones that are present at a ≥10% frequency in 1,165 exome sequences from tumors in The Cancer Genome Atlas. 86% of tumors across 12 cancer types had at least two clones. ITH in the morphology of nuclei was associated with genetic ITH (Spearman's correlation coefficient, ρ = 0.24-0.41; P < 0.001). Mutation of a driver gene that typically appears in smaller clones was a survival risk factor (hazard ratio (HR) = 2.15, 95% confidence interval (CI): 1.71-2.69). The risk of mortality also increased when >2 clones coexisted in the same tumor sample (HR = 1.49, 95% CI: 1.20-1.87). In two independent data sets, copy-number alterations affecting either <25% or >75% of a tumor's genome predicted reduced risk (HR = 0.15, 95% CI: 0.08-0.29). Mortality risk also declined when >4 clones coexisted in the sample, suggesting a trade-off between the costs and benefits of genomic instability. ITH and genomic instability thus have the potential to be useful measures that can universally be applied to all cancers.
Collapse
Affiliation(s)
- Noemi Andor
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Marnix Jansen
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li C Xia
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - C Athena Aktipis
- Center for Evolution and Cancer, University of California San Francisco, San Francisco, California, USA.,Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | - Claudia Petritsch
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA.,Brain Tumor Research Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Stanford Genome Technology Center, Stanford University, Palo Alto, California, USA
| | - Carlo C Maley
- Center for Evolution and Cancer, University of California San Francisco, San Francisco, California, USA.,Centre for Evolution and Cancer, Institute for Cancer Research, London, UK.,Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
161
|
Nawaz S, Yuan Y. Computational pathology: Exploring the spatial dimension of tumor ecology. Cancer Lett 2015; 380:296-303. [PMID: 26592351 DOI: 10.1016/j.canlet.2015.11.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023]
Abstract
Tumors are evolving ecosystems where cancer subclones and the microenvironment interact. This is analogous to interaction dynamics between species in their natural habitats, which is a prime area of study in ecology. Spatial statistics are frequently used in ecological studies to infer complex relations including predator-prey, resource dependency and co-evolution. Recently, the emerging field of computational pathology has enabled high-throughput spatial analysis by using image processing to identify different cell types and their locations within histological tumor samples. We discuss how these data may be analyzed with spatial statistics used in ecology to reveal patterns and advance our understanding of ecological interactions occurring among cancer cells and their microenvironment.
Collapse
Affiliation(s)
- Sidra Nawaz
- Centre for Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK; Centre for Evolution and Cancer, Institute of Cancer Research, London SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Yinyin Yuan
- Centre for Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK; Centre for Evolution and Cancer, Institute of Cancer Research, London SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
162
|
Frequent frameshift mutations in 2 mononucleotide repeats of RNF43 gene and its regional heterogeneity in gastric and colorectal cancers. Hum Pathol 2015; 46:1640-6. [DOI: 10.1016/j.humpath.2015.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/23/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023]
|
163
|
Combined bacterial and viral treatment: a novel anticancer strategy. Cent Eur J Immunol 2015; 40:366-72. [PMID: 26648783 PMCID: PMC4655389 DOI: 10.5114/ceji.2015.54601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
An idea for a new combination therapy will be described herein. It is a proposition to combine viral and bacterial anticancer therapies and make them fight cancer in concert. We analyzed biological anticancer therapies and found overlapping advantages and disadvantages which led us to the conclusion that the combination therapy has the potential to create a new therapeutic quality. It is surprising how many weaknesses of viral anticancer therapy are the strengths of bacterial anticancer therapies and the other way round. We review the facts behind this concept and try to assess its value. We propose a few strategies how to combine these two therapies but as far as the review can go, final answers will have to come from the experiments. This review is the first attempt to describe a new strategy and understand the means for this idea but also to raise new questions and discuss new ways to look at anti-cancer treatment.
Collapse
|
164
|
Intra-tumor heterogeneity of cancer cells and its implications for cancer treatment. Acta Pharmacol Sin 2015; 36:1219-27. [PMID: 26388155 PMCID: PMC4648179 DOI: 10.1038/aps.2015.92] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/06/2015] [Indexed: 02/06/2023]
Abstract
Recent studies have revealed extensive genetic and non-genetic variation across different geographical regions of a tumor or throughout different stages of tumor progression, which is referred to as intra-tumor heterogeneity. Several causes contribute to this phenomenon, including genomic instability, epigenetic alteration, plastic gene expression, signal transduction, and microenvironmental differences. These variables may affect key signaling pathways that regulate cancer cell growth, drive phenotypic diversity, and pose challenges to cancer treatment. Understanding the mechanisms underlying this heterogeneity will support the development of effective therapeutic strategies.
Collapse
|
165
|
Janiszewska M, Liu L, Almendro V, Kuang Y, Paweletz C, Sakr RA, Weigelt B, Hanker AB, Chandarlapaty S, King TA, Reis-Filho JS, Arteaga CL, Park SY, Michor F, Polyak K. In situ single-cell analysis identifies heterogeneity for PIK3CA mutation and HER2 amplification in HER2-positive breast cancer. Nat Genet 2015; 47:1212-9. [PMID: 26301495 PMCID: PMC4589505 DOI: 10.1038/ng.3391] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/31/2015] [Indexed: 12/19/2022]
Abstract
Detection of minor, genetically distinct subpopulations within tumors is a key challenge in cancer genomics. Here we report STAR-FISH (specific-to-allele PCR-FISH), a novel method for the combined detection of single-nucleotide and copy number alterations in single cells in intact archived tissues. Using this method, we assessed the clinical impact of changes in the frequency and topology of PIK3CA mutation and HER2 (ERBB2) amplification within HER2-positive breast cancer during neoadjuvant therapy. We found that these two genetic events are not always present in the same cells. Chemotherapy selects for PIK3CA-mutant cells, a minor subpopulation in nearly all treatment-naive samples, and modulates genetic diversity within tumors. Treatment-associated changes in the spatial distribution of cellular genetic diversity correlated with poor long-term outcome following adjuvant therapy with trastuzumab. Our findings support the use of in situ single cell-based methods in cancer genomics and imply that chemotherapy before HER2-targeted therapy may promote treatment resistance.
Collapse
Affiliation(s)
- Michalina Janiszewska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Lin Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Vanessa Almendro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yanan Kuang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Belfer Institute of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Cloud Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Belfer Institute of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rita A Sakr
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ariella B Hanker
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tari A King
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Carlos L Arteaga
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Medicine, Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
166
|
Böttger K, Hatzikirou H, Voss-Böhme A, Cavalcanti-Adam EA, Herrero MA, Deutsch A. An Emerging Allee Effect Is Critical for Tumor Initiation and Persistence. PLoS Comput Biol 2015; 11:e1004366. [PMID: 26335202 PMCID: PMC4559422 DOI: 10.1371/journal.pcbi.1004366] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 06/01/2015] [Indexed: 11/19/2022] Open
Abstract
Tumor cells develop different strategies to cope with changing microenvironmental conditions. A prominent example is the adaptive phenotypic switching between cell migration and proliferation. While it has been shown that the migration-proliferation plasticity influences tumor spread, it remains unclear how this particular phenotypic plasticity affects overall tumor growth, in particular initiation and persistence. To address this problem, we formulate and study a mathematical model of spatio-temporal tumor dynamics which incorporates the microenvironmental influence through a local cell density dependence. Our analysis reveals that two dynamic regimes can be distinguished. If cell motility is allowed to increase with local cell density, any tumor cell population will persist in time, irrespective of its initial size. On the contrary, if cell motility is assumed to decrease with respect to local cell density, any tumor population below a certain size threshold will eventually extinguish, a fact usually termed as Allee effect in ecology. These results suggest that strategies aimed at modulating migration are worth to be explored as alternatives to those mainly focused at keeping tumor proliferation under control. Controlling tumor growth remains a major medical challenge. Current clinical therapies focus on strategies to reduce tumor cell proliferation. However, during tumor progression, tumor cells may switch between proliferative and migratory behaviors, thereby allowing adaptation to microenvironmental changes that result in variations in local tumor cell density. We herein explore by means of a mathematical model the impact of migration-proliferation plasticity on tumor initiation and persistence. Our work suggests that small tumors can become extinct solely by their intrinsic cell population dynamics if cell motility decreases along with local cell density. In contrast, if cell motility increases with cell density, the tumor inevitably grows. Our model suggests that the regulation of cell migration plays a key role in tumor growth as a whole, making this feature a potential target for clinical studies.
Collapse
Affiliation(s)
- Katrin Böttger
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | | | - Anja Voss-Böhme
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
- Hochschule für Technik und Wirtschaft Dresden, Dresden, Germany
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Heidelberg, Germany
- Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Miguel A. Herrero
- Departamento de Matemática Aplicada, Facultad de Matemáticas, Universidad Complutense, Madrid, Spain
| | - Andreas Deutsch
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
167
|
Kolch W, Halasz M, Granovskaya M, Kholodenko BN. The dynamic control of signal transduction networks in cancer cells. Nat Rev Cancer 2015; 15:515-27. [PMID: 26289315 DOI: 10.1038/nrc3983] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is often considered a genetic disease. However, much of the enormous plasticity of cancer cells to evolve different phenotypes, to adapt to challenging microenvironments and to withstand therapeutic assaults is encoded by the structure and spatiotemporal dynamics of signal transduction networks. In this Review, we discuss recent concepts concerning how the rich signalling dynamics afforded by these networks are regulated and how they impinge on cancer cell proliferation, survival, invasiveness and drug resistance. Understanding this dynamic circuitry by mathematical modelling could pave the way to new therapeutic approaches and personalized treatments.
Collapse
Affiliation(s)
- Walter Kolch
- Systems Biology Ireland, University College Dublin
- Conway Institute of Biomolecular &Biomedical Research, University College Dublin
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Marina Granovskaya
- Roche Moscow Limited, Business Center Neglinnaya Plaza, Building 2, Trubnaya Square, 107031 Moscow, Russia
| | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin
- Conway Institute of Biomolecular &Biomedical Research, University College Dublin
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
168
|
Abstract
PURPOSE OF REVIEW To describe recent advances in the application of advanced genomic technologies towards the identification of biomarkers of prognosis and treatment response in breast cancer. RECENT FINDINGS Advances in high-throughput genomic profiling such as massively parallel sequencing have enabled researchers to catalogue the spectrum of somatic alterations in breast cancers. These tools also hold promise for precision medicine through accurate patient prognostication, stratification, and the dynamic monitoring of treatment response. For example, recent efforts have defined robust molecular subgroups of breast cancer and novel subtype-specific oncogenes. In addition, previously unappreciated activating mutations in human epidermal growth factor receptor 2 have been reported, suggesting new therapeutic opportunities. Genomic profiling of cell-free tumor DNA and circulating tumor cells has been used to monitor disease burden and the emergence of resistance, and such 'liquid biopsy' approaches may facilitate the early, noninvasive detection of aggressive disease. Finally, single-cell genomics is coming of age and will contribute to an understanding of breast cancer evolutionary dynamics. SUMMARY Here, we highlight recent studies that employ high-throughput genomic technologies in an effort to elucidate breast cancer biology, discover new therapeutic targets, improve prognostication and stratification, and discuss the implications for precision cancer medicine.
Collapse
|
169
|
Lazebnik Y. The shock of being united and symphiliosis. Another lesson from plants? Cell Cycle 2015; 13:2323-9. [PMID: 25483182 DOI: 10.4161/cc.29704] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Yuri Lazebnik
- a Yale Cardiovascular Research Center; New Haven, CT USA
| |
Collapse
|
170
|
Abstract
Although it is widely accepted that most cancers exhibit some degree of intratumour heterogeneity, we are far from understanding the dynamics that operate among subpopulations within tumours. There is growing evidence that cancer cells behave as communities, and increasing attention is now being directed towards the cooperative behaviour of subclones that can influence disease progression. As expected, these interactions can add a greater layer of complexity to therapeutic interventions in heterogeneous tumours, often leading to a poor prognosis. In this Review, we highlight studies that demonstrate such interactions in cancer and postulate ways to overcome them with better-designed therapeutic strategies.
Collapse
Affiliation(s)
- Doris P Tabassum
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA. [2] BBS Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kornelia Polyak
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA. [2] BBS Program, Harvard Medical School, Boston, Massachusetts 02115, USA. [3] Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA. [4] Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
171
|
Brock A, Krause S, Ingber DE. Control of cancer formation by intrinsic genetic noise and microenvironmental cues. Nat Rev Cancer 2015; 15:499-509. [PMID: 26156637 DOI: 10.1038/nrc3959] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Differentiation therapies that induce malignant cells to stop growing and revert to normal tissue-specific differentiated cell types are successful in the treatment of a few specific haematological tumours. However, this approach has not been widely applied to solid tumours because their developmental origins are less well understood. Recent advances suggest that understanding tumour cell plasticity and how intrinsic factors (such as genetic noise and microenvironmental signals, including physical cues from the extracellular matrix) govern cell state switches will help in the development of clinically relevant differentiation therapies for solid cancers.
Collapse
Affiliation(s)
- Amy Brock
- 1] Department of Biomedical Engineering, Institute for Cell and Molecular Biology, The University of Texas, Austin, Texas 78712, USA. [2]
| | - Silva Krause
- 1] Momenta Pharmaceuticals, Cambridge, Massachusetts 02142, USA. [2]
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, CLSB 5, Boston, Massachusetts 02115, USA
| |
Collapse
|
172
|
Budczies J, Bockmayr M, Denkert C, Klauschen F, Lennerz JK, Györffy B, Dietel M, Loibl S, Weichert W, Stenzinger A. Classical pathology and mutational load of breast cancer - integration of two worlds. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2015; 1:225-38. [PMID: 27499907 PMCID: PMC4939893 DOI: 10.1002/cjp2.25] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/13/2015] [Indexed: 12/18/2022]
Abstract
Breast cancer is a complex molecular disease comprising several biological subtypes. However, daily routine diagnosis is still based on a small set of well‐characterized clinico‐pathological variables. Here, we try to link the two worlds of surgical pathology and multilayered molecular profiling by analyzing the relationships between clinico‐pathological phenotypes and mutational loads of breast cancer. We evaluated the number of mutated genes with somatic non‐silent mutations in different subgroups of breast cancer based on clinico‐pathological, including immunohistochemical and tumour characteristics. The analysis was performed for a cohort of 687 primary breast cancer patients with mutational profiling, gene expression and clinico‐pathological data available from The Cancer Genome Atlas (TCGA) project. The number of mutated genes was strongly positively associated with higher tumour grade (p = 1.4e−14) and with the different immunohistochemical and PAM50 molecular subtypes of breast cancer (p = 1.4e−10 and p = 4.3e−10, respectively). We observed significant associations (|R| > 0.4) between the abundance of mutated genes and expression levels of genes related to proliferation in the overall cohort and hormone receptor positive cohort, including the Recurrence Score gene signature (e.g., MYBL2 and BIRC5). Specific mutated genes (TP53, NCOR1, NF1, PTPRD and RB1) were highly significantly associated with high loads of mutated genes. Multivariate analysis for overall survival (OS) revealed a worse survival for patients with high numbers of mutated genes (hazard ratio = 4.6, 95% CI: 1.0 – 20.0, p = 0.044). Here, we report a strong association of the number of mutated genes with immunohistochemical and PAM50 subtypes and tumour grade in breast cancer. We provide evidence that specific levels of the mutational load underlie different morphological and biological phenotypes, which collectively constitute the current basis of pathological diagnosis. Our study is a step towards genomics‐informed breast pathology and will provide a basis for future studies in this field bridging the gap between morphology, tumour biology and medical oncology.
Collapse
Affiliation(s)
- Jan Budczies
- Institute of Pathology, Charité University HospitalBerlinGermany; German Cancer Consortium (DKTK)BerlinGermany
| | - Michael Bockmayr
- Institute of Pathology, Charité University Hospital Berlin Germany
| | - Carsten Denkert
- Institute of Pathology, Charité University HospitalBerlinGermany; German Cancer Consortium (DKTK)BerlinGermany
| | | | - Jochen K Lennerz
- Massachusetts General Hospital/Harvard Medical School, Department of Pathology, Center for Integrated Diagnostics (CID) Boston Massachusetts
| | - Balázs Györffy
- MTA TTK Lendület Cancer Biomarker Research GroupBudapestHungary; 2nd Department of PediatricsSemmelweis UniversityBudapestHungary; MTA-SE Pediatrics and Nephrology Research GroupBudapestHungary
| | - Manfred Dietel
- Institute of Pathology, Charité University HospitalBerlinGermany; German Cancer Consortium (DKTK)BerlinGermany
| | | | - Wilko Weichert
- Institute of Pathology, University Hospital HeidelbergHeidelbergGermany; National Center for Tumor Diseases (NCT)HeidelbergGermany; German Cancer Consortium (DKTK)HeidelbergGermany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital HeidelbergHeidelbergGermany; Fellow of the NCT Heidelberg School of Oncology (NCT-HSO)HeidelbergGermany
| |
Collapse
|
173
|
Rovigatti U. Cancer modelling in the NGS era - Part I: Emerging technology and initial modelling. Crit Rev Oncol Hematol 2015; 96:274-307. [PMID: 26427785 DOI: 10.1016/j.critrevonc.2015.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 04/14/2015] [Accepted: 05/19/2015] [Indexed: 02/07/2023] Open
Abstract
It is today indisputable that great progresses have been made in our molecular understanding of cancer cells, but an effective implementation of such knowledge into dramatic cancer-cures is still belated and yet desperately needed. This review gives a snapshot at where we stand today in this search for cancer understanding and definitive treatments, how far we have progressed and what are the major obstacles we will have to overcome both technologically and for disease modelling. In the first part, promising 3rd/4th Generation Sequencing Technologies will be summarized (particularly IonTorrent and OxfordNanopore technologies). Cancer modelling will be then reviewed from its origin in XIX Century Germany to today's NGS applications for cancer understanding and therapeutic interventions. Developments after Molecular Biology revolution (1953) are discussed as successions of three phases. The first, PH1, labelled "Clonal Outgrowth" (from 1960s to mid 1980s) was characterized by discoveries in cytogenetics (Nowell, Rowley) and viral oncology (Dulbecco, Bishop, Varmus), which demonstrated clonality. Treatments were consequently dominated by a "cytotoxic eradication" strategy with chemotherapeutic agents. In PH2, (from the mid 1980s to our days) the description of cancer as "Gene Networks" led to targeted-gene-therapies (TGTs). TGTs are the focus of Section 3: in view of their apparent failing (Ephemeral Therapies), alternative strategies will be discussed in review part II (particularly cancer immunotherapy, CIT). Additional Pitfalls impinge on the concepts of tumour heterogeneity (inter/intra; ITH). The described pitfalls set the basis for a new phase, PH3, which is called "NGS Era" and will be also discussed with ten emerging cancer models in the Review 2nd part.
Collapse
Affiliation(s)
- Ugo Rovigatti
- University of Pisa Medical School, Oncology Department, via Roma 55, 56127 Pisa, Italy.
| |
Collapse
|
174
|
Yates LR, Gerstung M, Knappskog S, Desmedt C, Gundem G, Loo PV, Aas T, Alexandrov LB, Larsimont D, Davies H, Li Y, Ju YS, Ramakrishna M, Haugland HK, Lilleng PK, Nik-Zainal S, McLaren S, Butler A, Martin S, Glodzik D, Menzies A, Raine K, Hinton J, Jones D, Mudie LJ, Jiang B, Vincent D, Greene-Colozzi A, Adnet PY, Fatima A, Maetens M, Ignatiadis M, Stratton MR, Sotiriou C, Richardson AL, Lønning PE, Wedge DC, Campbell PJ. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med 2015; 21:751-9. [PMID: 26099045 PMCID: PMC4500826 DOI: 10.1038/nm.3886] [Citation(s) in RCA: 620] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022]
Abstract
The sequencing of cancer genomes may enable tailoring of therapeutics to the underlying biological abnormalities driving a particular patient's tumor. However, sequencing-based strategies rely heavily on representative sampling of tumors. To understand the subclonal structure of primary breast cancer, we applied whole-genome and targeted sequencing to multiple samples from each of 50 patients' tumors (303 samples in total). The extent of subclonal diversification varied among cases and followed spatial patterns. No strict temporal order was evident, with point mutations and rearrangements affecting the most common breast cancer genes, including PIK3CA, TP53, PTEN, BRCA2 and MYC, occurring early in some tumors and late in others. In 13 out of 50 cancers, potentially targetable mutations were subclonal. Landmarks of disease progression, such as resistance to chemotherapy and the acquisition of invasive or metastatic potential, arose within detectable subclones of antecedent lesions. These findings highlight the importance of including analyses of subclonal structure and tumor evolution in clinical trials of primary breast cancer.
Collapse
Affiliation(s)
- Lucy R Yates
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
- Department of Oncology, The University of Cambridge, Cambridge, UK
| | - Moritz Gerstung
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Christine Desmedt
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gunes Gundem
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Peter Van Loo
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
- Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Turid Aas
- Department of Surgery, Haukeland University Hospital, Bergen, Norway
| | - Ludmil B Alexandrov
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Denis Larsimont
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Helen Davies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Yilong Li
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Young Seok Ju
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | | | | | - Peer Kaare Lilleng
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- The Gade Laboratory for Pathology, Haukeland University Hospital, Bergen, Norway
| | | | - Stuart McLaren
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Adam Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Sancha Martin
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Dominic Glodzik
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Andrew Menzies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Jonathan Hinton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - David Jones
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Laura J Mudie
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Bing Jiang
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, USA
| | - Delphine Vincent
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Pierre-Yves Adnet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Aquila Fatima
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, USA
| | - Marion Maetens
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Michail Ignatiadis
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea L Richardson
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, USA
- Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - David C Wedge
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Peter J Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| |
Collapse
|
175
|
Menchón SA. The effect of intrinsic and acquired resistances on chemotherapy effectiveness. Acta Biotheor 2015; 63:113-27. [PMID: 25750013 DOI: 10.1007/s10441-015-9248-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 02/20/2015] [Indexed: 11/26/2022]
Abstract
Although chemotherapy is one of the most common treatments for cancer, it can be only partially successful. Drug resistance is the main cause of the failure of chemotherapy. In this work, we present a mathematical model to study the impact of both intrinsic (preexisting) and acquired (induced by the drugs) resistances on chemotherapy effectiveness. Our simulations show that intrinsic resistance could be as dangerous as acquired resistance. In particular, our simulations suggest that tumors composed by even a small fraction of intrinsically resistant cells may lead to an unsuccessful therapy very quickly. Our results emphasize the importance of monitoring both intrinsic and acquired resistances during treatment in order to succeed and the importance of doing more experimental and genetic research in order to develop a pretreatment clinical test to avoid intrinsic resistance.
Collapse
Affiliation(s)
- Silvia A Menchón
- IFEG-CONICET and FaMAF, Universidad Nacional de Córdoba, Medina Allende s/n, Ciudad Universitaria, 5000, Córdoba, Argentina,
| |
Collapse
|
176
|
Choi MR, Gwak M, Yoo NJ, Lee SH. Regional Bias of Intratumoral Genetic Heterogeneity of Apoptosis-Related Genes BAX, APAF1, and FLASH in Colon Cancers with High Microsatellite Instability. Dig Dis Sci 2015; 60:1674-9. [PMID: 25599959 DOI: 10.1007/s10620-014-3499-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/18/2014] [Indexed: 02/02/2023]
Abstract
BACKGROUND Apoptosis inactivation and intratumoral heterogeneity (ITH) are common features of cancers, including colorectal cancer (CRC). Inactivation of apoptosis prolongs cancer cell survival, and ITH may contribute to CRC progression. AIM To examine the presence and extent of mutational ITH in the pro-apoptotic genes APAF1, BAX, and FLASH and the association of mutational ITH with pathologic parameters of CRC. METHODS The ITH of mutations in the mononucleotide repeats of APAF1, BAX and FLASH in different tumors were analyzed in 16 cases of CRC with high microsatellite instability (MSI-H) and 41 cases of CRC with stable MSI/low MSI (MSS/MSI-L) by single-strand conformation polymorphism and DNA sequencing analyses. RESULTS Frameshift mutations of APAF1, BAX, and FLASH were identified in 19, 31, and 6 % of CRC with MSI-H, respectively, but also in cases of CRC with MSS/MSI-L. All but one CRC with a mutation (8/9) harbored regional ITH of the APAF1, BAX and FLASH frameshift mutations. ITH, however, was not associated with histopathologic features of CRC with MSI-H, suggesting that ITH might not be related to development of the MSI-H phenotype itself, but rather to disease progression. CONCLUSIONS Our results indicate that the APAF1, BAX, and FLASH genes not only harbor frameshift mutations but also demonstrate mutational ITH, which together might play a role in the tumorigenesis of CRC with MSI-H by affecting the apoptosis of cancer cells. Our data also suggest that multiregional mutation analysis is needed for a better evaluation of the mutation status in CRC.
Collapse
Affiliation(s)
- Mi Ryoung Choi
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul, 137-701, Korea
| | | | | | | |
Collapse
|
177
|
Turning the headlights on novel cancer biomarkers: Inspection of mechanics underlying intratumor heterogeneity. Mol Aspects Med 2015; 45:3-13. [PMID: 26024970 DOI: 10.1016/j.mam.2015.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/20/2015] [Indexed: 01/20/2023]
Abstract
Although the existence of intratumoral heterogeneity (ITH) in the expression of common biomarkers has been described by pathologists since the late 1890s, we have only recently begun to fathom the staggering extent and near ubiquity of this phenomenon. From the tumor's perspective, ITH provides a stabilizing diversity that allows for the evolution of aggressive cancer phenotypes. As the weight of the evidence correlating ITH to poor prognosis burgeons, it has become increasingly important to determine the mechanisms by which a tumor acquires ITH, find clinically-adaptable means to quantify ITH and design strategies to deal with the numerous profound clinical ramifications that ITH forces upon us. Elucidation of the drivers of ITH could enable development of novel biomarkers whose interrogation might permit quantitative evaluation of the ITH inherent in a tumor in order to predict the poor prognosis risk associated with that tumor. This review proposes centrosome amplification (CA), aided and abetted by centrosome clustering mechanisms, as a critical driver of chromosomal instability (CIN) that makes a key contribution to ITH generation. Herein we also evaluate how a tumor's inherent mitotic propensity, which reflects the cell cycling kinetics within the tumor's proliferative cells, functions as the indispensable engine underpinning CIN, and determines the rate of CIN. We thus expound how the forces of centrosome amplification and mitotic propensity collaborate to sculpt the genetic landscape of a tumor and spawn extensive subclonal diversity. As such, centrosome amplification and mitotic propensity profiles could serve as clinically facile and powerful prognostic biomarkers that would enable more accurate risk segmentation of patients and design of individualized therapies.
Collapse
|
178
|
Abstract
Hormone-receptor-positive breast cancer accounts for the majority-up to 80%-of all breast cancers. The evolution of breast cancer from early stage to the metastatic setting leads to increased heterogeneity, the occurrence of new mutations, and the development of treatment resistance representing a great challenge for management decisions. Unfortunately, little data exist to offer guidance in this context, and a reliance on traditional clinical parameters remains when deciding on optimal treatment. In advanced-stage oestrogen receptor-positive (ER+) disease, ongoing issues include the choice between endocrine therapy and chemotherapy, the appropriate sequence of treatment agents, and the incorporation of biological agents, such as everolimus, into the treatment armamentarium. In metastatic disease, repeated biopsies can help to reassess the receptor or genetic mutational status; however, the evidence to support this approach is limited. In this Review, we examine the current evidence that can guide treatment decisions in patients with advanced-stage ER+ breast cancer, discuss how to tackle these therapeutic challenges and provide suggestions for the optimal management of this patient population.
Collapse
|
179
|
Toss A, Cristofanilli M. Molecular characterization and targeted therapeutic approaches in breast cancer. Breast Cancer Res 2015; 17:60. [PMID: 25902832 PMCID: PMC4407294 DOI: 10.1186/s13058-015-0560-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite the wide improvements in breast cancer (BC) detection and adjuvant treatment, BC is still responsible for approximately 40,000 deaths annually in the United States. Novel biomarkers are fundamental to assist clinicians in BC detection, risk stratification, disease subtyping, prediction of treatment response, and surveillance, allowing a more tailored approach to therapy in both primary and metastatic settings. In primary BC, the development of molecular profiling techniques has added prognostic and predictive information to conventional biomarkers--estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Moreover, the application of next-generation sequencing and reverse-phase protein microarray methods in the metastatic setting holds the promise to further advance toward a personalized management of cancer. The improvement in our understanding on BC biology associated with the study of the genomic aberrations characterizing the most common molecular subtypes allows us to explore new targets for drug development. Finally, the integration of cancer stem cell-targeted therapies and immune therapies in future combination regimens increases our chances to successfully treat a larger proportion of women with more aggressive and resistant metastatic disease. This article reviews the current state of novel biological markers for BC, the evidence to demonstrate their clinical validity and utility, and the implication for therapeutic targeting.
Collapse
Affiliation(s)
- Angela Toss
- Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Via del Pozzo 71, Modena, 41125, Italy.
| | - Massimo Cristofanilli
- Department of Medical Oncology, Jefferson University Hospital, 1100 Walnut Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
180
|
Jonas O, Landry HM, Fuller JE, Santini JT, Baselga J, Tepper RI, Cima MJ, Langer R. An implantable microdevice to perform high-throughput in vivo drug sensitivity testing in tumors. Sci Transl Med 2015; 7:284ra57. [PMID: 25904741 PMCID: PMC4825177 DOI: 10.1126/scitranslmed.3010564] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Current anticancer chemotherapy relies on a limited set of in vitro or indirect prognostic markers of tumor response to available drugs. A more accurate analysis of drug sensitivity would involve studying tumor response in vivo. To this end, we have developed an implantable device that can perform drug sensitivity testing of several anticancer agents simultaneously inside the living tumor. The device contained reservoirs that released microdoses of single agents or drug combinations into spatially distinct regions of the tumor. The local drug concentrations were chosen to be representative of concentrations achieved during systemic treatment. Local efficacy and drug concentration profiles were evaluated for each drug or drug combination on the device, and the local efficacy was confirmed to be a predictor of systemic efficacy in vivo for multiple drugs and tumor models. Currently, up to 16 individual drugs or combinations can be assessed independently, without systemic drug exposure, through minimally invasive biopsy of a small region of a single tumor. This assay takes into consideration physiologic effects that contribute to drug response by allowing drugs to interact with the living tumor in its native microenvironment. Because these effects are crucial to predicting drug response, we envision that these devices will help identify optimal drug therapy before systemic treatment is initiated and could improve drug response prediction beyond the biomarkers and in vitro and ex vivo studies used today. These devices may also be used in clinical drug development to safely gather efficacy data on new compounds before pharmacological optimization.
Collapse
Affiliation(s)
- Oliver Jonas
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather M Landry
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason E Fuller
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Kibur Medical Inc., 29 Newbury Street, Suite 301, Boston, MA 02116, USA
| | - John T Santini
- Kibur Medical Inc., 29 Newbury Street, Suite 301, Boston, MA 02116, USA
| | - Jose Baselga
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Robert I Tepper
- Kibur Medical Inc., 29 Newbury Street, Suite 301, Boston, MA 02116, USA. Third Rock Ventures LLC, 29 Newbury Street, Boston, MA 02116, USA
| | - Michael J Cima
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Materials Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
181
|
Modelska A, Quattrone A, Re A. Molecular portraits: the evolution of the concept of transcriptome-based cancer signatures. Brief Bioinform 2015; 16:1000-7. [PMID: 25832647 PMCID: PMC4652618 DOI: 10.1093/bib/bbv013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Indexed: 12/13/2022] Open
Abstract
Cancer results from dysregulation of multiple steps of gene expression programs. We review how transcriptome profiling has been widely explored for cancer classification and biomarker discovery but resulted in limited clinical impact. Therefore, we discuss alternative and complementary omics approaches.
Collapse
|
182
|
Yang YC, Li XP. Clinical significance of intratumor heterogeneity for gynecological carcinoma. Chronic Dis Transl Med 2015; 1:14-17. [PMID: 29062982 PMCID: PMC5643778 DOI: 10.1016/j.cdtm.2015.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Indexed: 01/23/2023] Open
Affiliation(s)
- Ying-Chao Yang
- Department of Gynaecology, Peking University People's Hospital, Beijing 100044, China
| | - Xiao-Ping Li
- Department of Gynaecology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
183
|
Zhang M, Tsimelzon A, Chang CH, Fan C, Wolff A, Perou CM, Hilsenbeck SG, Rosen JM. Intratumoral heterogeneity in a Trp53-null mouse model of human breast cancer. Cancer Discov 2015; 5:520-33. [PMID: 25735774 DOI: 10.1158/2159-8290.cd-14-1101] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/20/2015] [Indexed: 12/24/2022]
Abstract
UNLABELLED Intratumoral heterogeneity correlates with clinical outcome and reflects the cellular complexity and dynamics within a tumor. Such heterogeneity is thought to contribute to radio- and chemoresistance because many treatments may target only certain tumor cell subpopulations. A better understanding of the functional interactions between various subpopulations of cells, therefore, may help in the development of effective cancer treatments. We identified a unique subpopulation of tumor cells expressing mesenchymal-like markers in a Trp53-null mouse model of basal-like breast cancer using fluorescence-activated cell sorting and microarray analysis. Both in vitro and in vivo experiments revealed the existence of cross-talk between these "mesenchymal-like" cells and tumor-initiating cells. Knockdown of genes encoding ligands upregulated in the mesenchymal cells and their corresponding receptors in the tumor-initiating cells resulted in reduced tumorigenicity and increased tumor latency. These studies illustrate the non-cell-autonomous properties and importance of cooperativity between tumor subpopulations. SIGNIFICANCE Intratumoral heterogeneity has been considered one important factor in assessing a patient's initial response to treatment and selecting drug regimens to effectively increase tumor response rate. Elucidating the functional interactions between various subpopulations of tumor cells will help provide important new insights in understanding treatment response and tumor progression.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Chi-Hsuan Chang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew Wolff
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
184
|
Rello-Varona S, Herrero-Martín D, López-Alemany R, Muñoz-Pinedo C, Tirado OM. “(Not) All (Dead) Things Share the Same Breath”: Identification of Cell Death Mechanisms in Anticancer Therapy. Cancer Res 2015; 75:913-7. [DOI: 10.1158/0008-5472.can-14-3494] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
185
|
Yuan K, Sakoparnig T, Markowetz F, Beerenwinkel N. BitPhylogeny: a probabilistic framework for reconstructing intra-tumor phylogenies. Genome Biol 2015; 16:36. [PMID: 25786108 PMCID: PMC4359483 DOI: 10.1186/s13059-015-0592-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/21/2015] [Indexed: 11/28/2022] Open
Abstract
Cancer has long been understood as a somatic evolutionary process, but many details of tumor progression remain elusive. Here, we present BitPhylogenyBitPhylogeny, a probabilistic framework to reconstruct intra-tumor evolutionary pathways. Using a full Bayesian approach, we jointly estimate the number and composition of clones in the sample as well as the most likely tree connecting them. We validate our approach in the controlled setting of a simulation study and compare it against several competing methods. In two case studies, we demonstrate how BitPhylogeny BitPhylogeny reconstructs tumor phylogenies from methylation patterns in colon cancer and from single-cell exomes in myeloproliferative neoplasm.
Collapse
Affiliation(s)
- Ke Yuan
- />University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Thomas Sakoparnig
- />Department of Biosystems Science and Engineering, ETH Zurich, Basel Switzerland
- />SIB Swiss Institute of Bioinformatics, Basel, Switzerland
- />Current address: Biozentrum, University of Basel, Basel, Switzerland
| | - Florian Markowetz
- />University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Niko Beerenwinkel
- />Department of Biosystems Science and Engineering, ETH Zurich, Basel Switzerland
- />SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
186
|
Sottoriva A, Kang H, Ma Z, Graham TA, Salomon MP, Zhao J, Marjoram P, Siegmund K, Press MF, Shibata D, Curtis C. A Big Bang model of human colorectal tumor growth. Nat Genet 2015; 47:209-16. [PMID: 25665006 PMCID: PMC4575589 DOI: 10.1038/ng.3214] [Citation(s) in RCA: 754] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/12/2015] [Indexed: 12/12/2022]
Abstract
What happens in the early, still undetectable human malignancy is unknown because direct observations are impractical. Here we present and validate a “Big Bang” model, whereby tumors grow predominantly as a single expansion producing numerous intermixed sub-clones that are not subject to stringent selection, and where both public (clonal) and most detectable private (subclonal) alterations arise early during growth. Genomic profiling of 349 individual glands from 15 colorectal tumors revealed the absence of selective sweeps, uniformly high intra-tumor heterogeneity (ITH), and sub-clone mixing in distant regions, as postulated by our model. We also verified the prediction that most detectable ITH originates from early private alterations, and not from later clonal expansions, thus exposing the profile of the primordial tumor. Moreover, some tumors appear born-to-be-bad, with sub-clone mixing indicative of early malignant potential. This new model provides a quantitative framework to interpret tumor growth dynamics and the origins of ITH with significant clinical implications.
Collapse
Affiliation(s)
- Andrea Sottoriva
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Haeyoun Kang
- 1] Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA. [2] Department of Pathology, CHA University, Seongnam-si, South Korea
| | - Zhicheng Ma
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Trevor A Graham
- 1] Center for Evolution and Cancer, University of California, San Francisco, San Francisco, California, USA. [2] Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Matthew P Salomon
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Junsong Zhao
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Paul Marjoram
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kimberly Siegmund
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Michael F Press
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Darryl Shibata
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Christina Curtis
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
187
|
Guldner IH, Zhang S. A journey to uncharted territory: new technical frontiers in studying tumor-stromal cell interactions. Integr Biol (Camb) 2015; 7:153-61. [PMID: 25500646 PMCID: PMC4324098 DOI: 10.1039/c4ib00192c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The crosstalk between tumor cells and cells of the tumor stroma dictate malignant progression and represent an intriguing and viable anticancer therapeutic target. The successful development of therapeutics targeting tumor-stroma interactions is tied to the insight provided by basic research on such crosstalk. Tumor-stroma interactions can be transient and dynamic, and they occur within defined spatiotemporal contexts among genetically and compositionally heterogeneous populations of cells, yet methods currently applied to study the said crosstalk do not sufficiently address these features. Emerging imaging and genetic methods, however, can overcome limitations of traditional approaches and provide unprecedented insight into tumor-stroma crosstalk with unparalleled accuracy. The comprehensive data obtained by applying emerging methods will require processing and analysis by multidisciplinary teams, but the efforts will ultimately rejuvenate hope in developing novel therapies against pro-tumorigenic tumor-stroma crosstalk.
Collapse
Affiliation(s)
- Ian H Guldner
- Department of Biological Science, Harper Cancer Research Institute, University of Notre Dame, A130 Harper Hall, Notre Dame, IN 46556, USA.
| | | |
Collapse
|
188
|
Walsh AJ, Skala MC. Optical metabolic imaging quantifies heterogeneous cell populations. BIOMEDICAL OPTICS EXPRESS 2015; 6:559-73. [PMID: 25780745 PMCID: PMC4354590 DOI: 10.1364/boe.6.000559] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 05/20/2023]
Abstract
The genetic and phenotypic heterogeneity of cancers can contribute to tumor aggressiveness, invasion, and resistance to therapy. Fluorescence imaging occupies a unique niche to investigate tumor heterogeneity due to its high resolution and molecular specificity. Here, heterogeneous populations are identified and quantified by combined optical metabolic imaging and subpopulation analysis (OMI-SPA). OMI probes the fluorescence intensities and lifetimes of metabolic enzymes in cells to provide images of cellular metabolism, and SPA models cell populations as mixed Gaussian distributions to identify cell subpopulations. In this study, OMI-SPA is characterized by simulation experiments and validated with cell experiments. To generate heterogeneous populations, two breast cancer cell lines, SKBr3 and MDA-MB-231, were co-cultured at varying proportions. OMI-SPA correctly identifies two populations with minimal mean and proportion error using the optical redox ratio (fluorescence intensity of NAD(P)H divided by the intensity of FAD), mean NAD(P)H fluorescence lifetime, and OMI index. Simulation experiments characterized the relationships between sample size, data standard deviation, and subpopulation mean separation distance required for OMI-SPA to identify subpopulations.
Collapse
|
189
|
Frameshift mutations of TAF1C gene, a core component for transcription by RNA polymerase I, and its regional heterogeneity in gastric and colorectal cancers. Pathology 2015; 47:101-4. [DOI: 10.1097/pat.0000000000000212] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
190
|
Ecker S, Pancaldi V, Rico D, Valencia A. Higher gene expression variability in the more aggressive subtype of chronic lymphocytic leukemia. Genome Med 2015; 7:8. [PMID: 25632304 PMCID: PMC4308895 DOI: 10.1186/s13073-014-0125-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022] Open
Abstract
Background Chronic lymphocytic leukemia (CLL) presents two subtypes which have drastically different clinical outcomes, IgVH mutated (M-CLL) and IgVH unmutated (U-CLL). So far, these two subtypes are not associated to clear differences in gene expression profiles. Interestingly, recent results have highlighted important roles for heterogeneity, both at the genetic and at the epigenetic level in CLL progression. Methods We analyzed gene expression data of two large cohorts of CLL patients and quantified expression variability across individuals to investigate differences between the two subtypes using different measures and statistical tests. Functional significance was explored by pathway enrichment and network analyses. Furthermore, we implemented a random forest approach based on expression variability to classify patients into disease subtypes. Results We found that U-CLL, the more aggressive type of the disease, shows significantly increased variability of gene expression across patients and that, overall, genes that show higher variability in the aggressive subtype are related to cell cycle, development and inter-cellular communication. These functions indicate a potential relation between gene expression variability and the faster progression of this CLL subtype. Finally, a classifier based on gene expression variability was able to correctly predict the disease subtype of CLL patients. Conclusions There are strong relations between gene expression variability and disease subtype linking significantly increased expression variability to phenotypes such as aggressiveness and resistance to therapy in CLL. Electronic supplementary material The online version of this article (doi:10.1186/s13073-014-0125-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simone Ecker
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Vera Pancaldi
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Daniel Rico
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Alfonso Valencia
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| |
Collapse
|
191
|
Beerenwinkel N, Schwarz RF, Gerstung M, Markowetz F. Cancer evolution: mathematical models and computational inference. Syst Biol 2015; 64:e1-25. [PMID: 25293804 PMCID: PMC4265145 DOI: 10.1093/sysbio/syu081] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 09/26/2014] [Indexed: 12/12/2022] Open
Abstract
Cancer is a somatic evolutionary process characterized by the accumulation of mutations, which contribute to tumor growth, clinical progression, immune escape, and drug resistance development. Evolutionary theory can be used to analyze the dynamics of tumor cell populations and to make inference about the evolutionary history of a tumor from molecular data. We review recent approaches to modeling the evolution of cancer, including population dynamics models of tumor initiation and progression, phylogenetic methods to model the evolutionary relationship between tumor subclones, and probabilistic graphical models to describe dependencies among mutations. Evolutionary modeling helps to understand how tumors arise and will also play an increasingly important prognostic role in predicting disease progression and the outcome of medical interventions, such as targeted therapy.
Collapse
Affiliation(s)
- Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB20RE, United Kingdom Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB20RE, United Kingdom
| | - Roland F Schwarz
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB20RE, United Kingdom
| | - Moritz Gerstung
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB20RE, United Kingdom
| | - Florian Markowetz
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB20RE, United Kingdom
| |
Collapse
|
192
|
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]
|
193
|
Thompson AM, Gansen A, Paguirigan AL, Kreutz JE, Radich JP, Chiu DT. Self-digitization microfluidic chip for absolute quantification of mRNA in single cells. Anal Chem 2014; 86:12308-14. [PMID: 25390242 PMCID: PMC4270397 DOI: 10.1021/ac5035924] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
![]()
Quantification
of mRNA in single cells provides direct insight
into how intercellular heterogeneity plays a role in disease progression
and outcomes. Quantitative polymerase chain reaction (qPCR), the current
gold standard for evaluating gene expression, is insufficient for
providing absolute measurement of single-cell mRNA transcript abundance.
Challenges include difficulties in handling small sample volumes and
the high variability in measurements. Microfluidic digital PCR provides
far better sensitivity for minute quantities of genetic material,
but the typical format of this assay does not allow for counting of
the absolute number of mRNA transcripts samples taken from single
cells. Furthermore, a large fraction of the sample is often lost during
sample handling in microfluidic digital PCR. Here, we report the absolute
quantification of single-cell mRNA transcripts by digital, one-step
reverse transcription PCR in a simple microfluidic array device called
the self-digitization (SD) chip. By performing the reverse transcription
step in digitized volumes, we find that the assay exhibits a linear
signal across a wide range of total RNA concentrations and agrees
well with standard curve qPCR. The SD chip is found to digitize a
high percentage (86.7%) of the sample for single-cell experiments.
Moreover, quantification of transferrin receptor mRNA in single cells
agrees well with single-molecule fluorescence in situ hybridization
experiments. The SD platform for absolute quantification of single-cell
mRNA can be optimized for other genes and may be useful as an independent
control method for the validation of mRNA quantification techniques.
Collapse
Affiliation(s)
- Alison M Thompson
- Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | | | | | | | | | | |
Collapse
|
194
|
Carroll SP, Jørgensen PS, Kinnison MT, Bergstrom CT, Denison RF, Gluckman P, Smith TB, Strauss SY, Tabashnik BE. Applying evolutionary biology to address global challenges. Science 2014; 346:1245993. [PMID: 25213376 PMCID: PMC4245030 DOI: 10.1126/science.1245993] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two categories of evolutionary challenges result from escalating human impacts on the planet. The first arises from cancers, pathogens, and pests that evolve too quickly and the second, from the inability of many valued species to adapt quickly enough. Applied evolutionary biology provides a suite of strategies to address these global challenges that threaten human health, food security, and biodiversity. This Review highlights both progress and gaps in genetic, developmental, and environmental manipulations across the life sciences that either target the rate and direction of evolution or reduce the mismatch between organisms and human-altered environments. Increased development and application of these underused tools will be vital in meeting current and future targets for sustainable development.
Collapse
Affiliation(s)
- Scott P Carroll
- Department of Entomology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA. Institute for Contemporary Evolution, Davis, CA 95616, USA.
| | - Peter Søgaard Jørgensen
- Center for Macroecology, Evolution and Climate, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark. Center for Macroecology, Evolution and Climate, Natural History Museum of Denmark, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Michael T Kinnison
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA
| | - Carl T Bergstrom
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - R Ford Denison
- Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN 55108, USA
| | - Peter Gluckman
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Thomas B Smith
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA. Center for Tropical Research, Institute of the Environment and Sustainability, University of California, Los Angeles, 619 Charles E. Young Drive East, Los Angeles, 90095-1496, CA
| | - Sharon Y Strauss
- Department of Evolution and Ecology and Center for Population Biology, University of California, Davis, One Shields Avenue, CA 95616, USA
| | - Bruce E Tabashnik
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
195
|
Abstract
In this issue of Cell Reports, Almendro et al. report one of the first comprehensive studies on the intratumor heterogeneity of cell phenotypes and genotypes before and after chemotherapy in breast cancer. These data challenge the concept of genetic population bottlenecks and suggest that cellular phenotypes play an important role in developing resistance to therapy.
Collapse
Affiliation(s)
- Nicholas E Navin
- Department of Genetics, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
196
|
Jiang T, Shi W, Natowicz R, Ononye SN, Wali VB, Kluger Y, Pusztai L, Hatzis C. Statistical measures of transcriptional diversity capture genomic heterogeneity of cancer. BMC Genomics 2014; 15:876. [PMID: 25294321 PMCID: PMC4197225 DOI: 10.1186/1471-2164-15-876] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/24/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Molecular heterogeneity of tumors suggests the presence of multiple different subclones that may limit response to targeted therapies and contribute to acquisition of drug resistance, but its quantification has remained challenging. RESULTS We performed simulations to evaluate statistical measures that best capture the molecular diversity within a group of tumors for either continuous (gene expression) or discrete (mutations, copy number alterations) molecular data. Dispersion based metrics in the principal component space best captured the underlying heterogeneity. To demonstrate utility of these measures, we characterized the diversity in transcriptional and genomic profiles of different breast tumor subtypes, and showed that basal-like or triple-negative breast cancers (TNBC) are significantly more heterogeneous molecularly than other subtypes. Our analysis also suggests that transcriptional diversity is a global characteristic of the tumors observed across the majority of molecular pathways. Among basal-like tumors, those that were resistant to multi-agent chemotherapy showed greater transcriptional diversity compared to chemotherapy-sensitive tumors, suggesting that potentially multiple mechanisms may be contributing to chemotherapy resistance. CONCLUSIONS We proposed and validated measures of transcriptional and genomic diversity that can quantify the molecular diversity of tumors. We applied the new measures to genomic data from breast tumors and demonstrated that basal-like breast cancers are significantly more diverse than other breast cancers. The observation that chemo-resistant tumors are significantly more diverse molecularly than chemosensitive tumors implies that multiple resistance mechanisms may be active, thus limiting the sensitivity and accuracy of predictive markers of chemotherapy response.
Collapse
Affiliation(s)
- Tingting Jiang
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - Weiwei Shi
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - René Natowicz
- />Computer Science Department, Universite Paris-Est, Paris, France
| | - Sophia N Ononye
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - Vikram B Wali
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - Yuval Kluger
- />Department of Pathology, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - Lajos Pusztai
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| | - Christos Hatzis
- />Department of Internal Medicine, Yale School of Medicine, Yale Cancer Center, New Haven, Connecticut USA
| |
Collapse
|
197
|
Du W, Elemento O. Cancer systems biology: embracing complexity to develop better anticancer therapeutic strategies. Oncogene 2014; 34:3215-25. [PMID: 25220419 DOI: 10.1038/onc.2014.291] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 12/20/2022]
Abstract
The transformation of normal cells into cancer cells and maintenance of the malignant state and phenotypes are associated with genetic and epigenetic deregulations, altered cellular signaling responses and aberrant interactions with the microenvironment. These alterations are constantly evolving as tumor cells face changing selective pressures induced by the cells themselves, the microenvironment and drug treatments. Tumors are also complex ecosystems where different, sometime heterogeneous, subclonal tumor populations and a variety of nontumor cells coexist in a constantly evolving manner. The interactions between molecules and between cells that arise as a result of these alterations and ecosystems are even more complex. The cancer research community is increasingly embracing this complexity and adopting a combination of systems biology methods and integrated analyses to understand and predictively model the activity of cancer cells. Systems biology approaches are helping to understand the mechanisms of tumor progression and design more effective cancer therapies. These approaches work in tandem with rapid technological advancements that enable data acquisition on a broader scale, with finer accuracy, higher dimensionality and higher throughput than ever. Using such data, computational and mathematical models help identify key deregulated functions and processes, establish predictive biomarkers and optimize therapeutic strategies. Moving forward, implementing patient-specific computational and mathematical models of cancer will significantly improve the specificity and efficacy of targeted therapy, and will accelerate the adoption of personalized and precision cancer medicine.
Collapse
Affiliation(s)
- W Du
- Laboratory of Cancer Systems Biology, Sandra and Edward Meyer Cancer Center, Department of Physiology and Biophysics, Institute for Computational Biomedicine and Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - O Elemento
- Laboratory of Cancer Systems Biology, Sandra and Edward Meyer Cancer Center, Department of Physiology and Biophysics, Institute for Computational Biomedicine and Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
198
|
Burrell RA, Swanton C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol Oncol 2014; 8:1095-111. [PMID: 25087573 PMCID: PMC5528620 DOI: 10.1016/j.molonc.2014.06.005] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022] Open
Abstract
Cancer drug resistance is a major problem, with the majority of patients with metastatic disease ultimately developing multidrug resistance and succumbing to their disease. Our understanding of molecular events underpinning treatment failure has been enhanced by new genomic technologies and pre-clinical studies. Intratumour genetic heterogeneity (ITH) is a prominent contributor to therapeutic failure, and it is becoming increasingly apparent that individual tumours may achieve resistance via multiple routes simultaneously - termed polyclonal resistance. Efforts to target single resistance mechanisms to overcome therapeutic failure may therefore yield only limited success. Clinical studies with sequential analysis of tumour material are needed to enhance our understanding of inter-clonal functional relationships and tumour evolution during therapy, and to improve drug development strategies in cancer medicine.
Collapse
Affiliation(s)
- Rebecca A Burrell
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3L7, UK; UCL Cancer Institute, Paul O'Gorman Building University College London, 72 Huntley Street, London WC1E 6DD, UK.
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3L7, UK; UCL Cancer Institute, Paul O'Gorman Building University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
199
|
Abstract
The study of single cancer cells has transformed from qualitative microscopic images to quantitative genomic datasets. This paradigm shift has been fueled by the development of single-cell sequencing technologies, which provide a powerful new approach to study complex biological processes in human cancers.
Collapse
|
200
|
Trinh A, Rye IH, Almendro V, Helland Å, Russnes HG, Markowetz F. GoIFISH: a system for the quantification of single cell heterogeneity from IFISH images. Genome Biol 2014; 15:442. [PMID: 25168174 PMCID: PMC4167144 DOI: 10.1186/s13059-014-0442-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/15/2014] [Indexed: 11/23/2022] Open
Abstract
Molecular analysis has revealed extensive intra-tumor heterogeneity in human cancer samples, but cannot identify cell-to-cell variations within the tissue microenvironment. In contrast, in situ analysis can identify genetic aberrations in phenotypically defined cell subpopulations while preserving tissue-context specificity. GoIFISHGoIFISH is a widely applicable, user-friendly system tailored for the objective and semi-automated visualization, detection and quantification of genomic alterations and protein expression obtained from fluorescence in situ analysis. In a sample set of HER2-positive breast cancers GoIFISHGoIFISH is highly robust in visual analysis and its accuracy compares favorably to other leading image analysis methods. GoIFISHGoIFISH is freely available at www.sourceforge.net/projects/goifish/.
Collapse
Affiliation(s)
- Anne Trinh
- />University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, CB2 0RE Cambridge UK
| | - Inga H Rye
- />Department of Genetics, Institute for Cancer Research, Postboks 4950 Nydalen, 0424 Oslo Norway
- />K. G. Jebsen Centre for Breast Cancer Research, University of Oslo, Postboks 4950 Nydalen, 0424 Oslo Norway
| | - Vanessa Almendro
- />Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, US
- />Harvard Medical School, Boston, US
| | - Åslaug Helland
- />Department of Genetics, Institute for Cancer Research, Postboks 4950 Nydalen, 0424 Oslo Norway
- />K. G. Jebsen Centre for Breast Cancer Research, University of Oslo, Postboks 4950 Nydalen, 0424 Oslo Norway
- />Department of Cancer treatment, Oslo University Hospital, Postboks 4950 Nydalen0424 Oslo, Norway
| | - Hege G Russnes
- />Department of Genetics, Institute for Cancer Research, Postboks 4950 Nydalen, 0424 Oslo Norway
- />K. G. Jebsen Centre for Breast Cancer Research, University of Oslo, Postboks 4950 Nydalen, 0424 Oslo Norway
- />Department of Pathology, Oslo University Hospital, Postboks 4950 Nydalen, 0424 Oslo, Norway
| | - Florian Markowetz
- />University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, CB2 0RE Cambridge UK
| |
Collapse
|