1851
|
Neelakantan D, Drasin DJ, Ford HL. Intratumoral heterogeneity: Clonal cooperation in epithelial-to-mesenchymal transition and metastasis. Cell Adh Migr 2014; 9:265-76. [PMID: 25482627 DOI: 10.4161/19336918.2014.972761] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although phenotypic intratumoral heterogeneity was first described many decades ago, the advent of next-generation sequencing has provided conclusive evidence that in addition to phenotypic diversity, significant genotypic diversity exists within tumors. Tumor heterogeneity likely arises both from clonal expansions, as well as from differentiation hierarchies existent in the tumor, such as that established by cancer stem cells (CSCs) and non-CSCs. These differentiation hierarchies may arise due to genetic mutations, epigenetic alterations, or microenvironmental influences. An additional differentiation hierarchy within epithelial tumors may arise when only a few tumor cells trans-differentiate into mesenchymal-like cells, a process known as epithelial-to-mesenchymal transition (EMT). Again, this process can be influenced by both genetic and non-genetic factors. In this review we discuss the evidence for clonal interaction and cooperation for tumor maintenance and progression, particularly with respect to EMT, and further address the far-reaching effects that tumor heterogeneity may have on cancer therapy.
Collapse
Key Words
- CLL, chronic lymphoblastic leukemia
- CSC, cancer stem cell
- EMP, epithelial-mesenchymal plasticity
- EMT, epithelial-to-mesenchymal transition
- GFP, green fluorescent protein.
- MET, mesenchymal-to-epithelial transition
- MMTV, mouse mammary tumor virus
- NGS, next generation sequencing
- OxR, oxaliplatin resistant
- SCLC, small cell lung cancer
- TGF-β, transforming growth factor-β
- cancer stem cells/CSCs
- clonal evolution
- epithelial-mesenchymal transition (EMT)
- hPDGF human platelet-derived growth factor
- intratumoral heterogeneity
- metastasis
- miRNA, microRNA
- non-cell autonomous
- tumor microenvironment
Collapse
Affiliation(s)
- Deepika Neelakantan
- a Department of Pharmacology ; University of Colorado; School of Medicine ; Aurora, CO USA
| | | | | |
Collapse
|
1852
|
Wheeler SE, Clark AM, Taylor DP, Young CL, Pillai VC, Stolz DB, Venkataramanan R, Lauffenburger D, Griffith L, Wells A. Spontaneous dormancy of metastatic breast cancer cells in an all human liver microphysiologic system. Br J Cancer 2014; 111:2342-50. [PMID: 25314052 PMCID: PMC4264444 DOI: 10.1038/bjc.2014.533] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/08/2014] [Accepted: 09/11/2014] [Indexed: 12/25/2022] Open
Abstract
Background: Metastatic outgrowth in breast cancer can occur years after a seeming cure. Existing model systems of dormancy are limited as they do not recapitulate human metastatic dormancy without exogenous manipulations and are unable to query early events of micrometastases. Methods: Here, we describe a human ex vivo hepatic microphysiologic system. The system is established with fresh human hepatocytes and non-parenchymal cells (NPCs) creating a microenvironment into which breast cancer cells (MCF7 and MDA-MB-231) are added. Results: The hepatic tissue maintains function through 15 days as verified by liver-specific protein production and drug metabolism assays. The NPCs form an integral part of the hepatic niche, demonstrated within the system through their participation in differential signalling cascades and cancer cell outcomes. Breast cancer cells intercalate into the hepatic niche without interfering with hepatocyte function. Examination of cancer cells demonstrated that a significant subset enter a quiescent state of dormancy as shown by lack of cell cycling (EdU− or Ki67−). The presence of NPCs altered the cancer cell fraction entering quiescence, and lead to differential cytokine profiles in the microenvironment effluent. Conclusions: These findings establish the liver microphysiologic system as a relevant model for the study of breast cancer metastases and entry into dormancy.
Collapse
Affiliation(s)
- S E Wheeler
- Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA
| | - A M Clark
- Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA
| | - D P Taylor
- 1] Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA [2] Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - C L Young
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - V C Pillai
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - D B Stolz
- 1] Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA [2] Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA [3] McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA [4] University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - R Venkataramanan
- 1] Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA [2] Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - D Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - L Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A Wells
- 1] Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, USA [2] Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA [3] McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA [4] University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA [5] Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
1853
|
Nielsen TK, Højgaard M, Andersen JT, Poulsen HE, Lykkesfeldt J, Mikines KJ. Elimination of ascorbic acid after high-dose infusion in prostate cancer patients: a pharmacokinetic evaluation. Basic Clin Pharmacol Toxicol 2014; 116:343-8. [PMID: 25220574 DOI: 10.1111/bcpt.12323] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 01/14/2023]
Abstract
Treatment with high-dose intravenous (IV) ascorbic acid (AA) is used in complementary and alternative medicine for various conditions including cancer. Cytotoxicity to cancer cell lines has been observed with millimolar concentrations of AA. Little is known about the pharmacokinetics of high-dose IV AA. The purpose of this study was to assess the basic kinetic variables in human beings over a relevant AA dosing interval for proper design of future clinical trials. Ten patients with metastatic prostate cancer were treated for 4 weeks with fixed AA doses of 5, 30 and 60 g. AA was measured consecutively in plasma and indicated first-order elimination kinetics throughout the dosing range with supra-physiological concentrations. The target dose of 60 g AA IV produced a peak plasma AA concentration of 20.3 mM. Elimination half-life was 1.87 hr (mean, S.D. ± 0.40), volume of distribution 0.19 L/kg (S.D. ±0.05) and clearance rate 6.02 L/hr (100 mL/min). No differences in pharmacokinetic parameters were observed between weeks/doses. A relatively fast first-order elimination with half-life of about 2 hr makes it impossible to maintain AA concentrations in the potential cytotoxic range after infusion stop in prostate cancer patients with normal kidney function. We propose a regimen with a bolus loading followed by a maintenance infusion based on the calculated clearance.
Collapse
Affiliation(s)
- Torben K Nielsen
- Department of Urology, Copenhagen University Hospital Herlev, Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
1854
|
Methods to study the tumor microenvironment under controlled oxygen conditions. Trends Biotechnol 2014; 32:556-563. [PMID: 25282035 DOI: 10.1016/j.tibtech.2014.09.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/25/2014] [Accepted: 09/09/2014] [Indexed: 12/16/2022]
Abstract
The tumor microenvironment (TME) is a complex heterogeneous assembly composed of a variety of cell types and physical features. One such feature, hypoxia, is associated with metabolic reprogramming, the epithelial-mesenchymal transition, and therapeutic resistance. Many questions remain regarding the effects of hypoxia on these outcomes; however, only a few experimental methods enable both precise control over oxygen concentration and real-time imaging of cell behavior. Recent efforts with microfluidic platforms offer a promising solution to these limitations. In this review, we discuss conventional methods and tools used to control oxygen concentration for cell studies, and then highlight recent advances in microfluidic-based approaches for controlling oxygen in engineered platforms.
Collapse
|
1855
|
Mechanism-based cancer therapy: resistance to therapy, therapy for resistance. Oncogene 2014; 34:3617-26. [PMID: 25263438 DOI: 10.1038/onc.2014.314] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 08/19/2014] [Accepted: 08/19/2014] [Indexed: 12/18/2022]
Abstract
The introduction of targeted therapy promised personalized and efficacious cancer treatments. However, although some targeted therapies have undoubtedly improved prognosis and outcome for specific cancer patients, the recurrent problem of therapeutic resistance subdues present revolutionary claims in this field. The plasticity of tumor cells leads to the development of drug resistance by distinct mechanisms: (1) mutations in the target, (2) reactivation of the targeted pathway, (3) hyperactivation of alternative pathways and (4) cross-talk with the microenvironment. Moreover, the intra-tumor heterogeneity of most tumors can also limit therapeutic response. Interestingly, the early identification of some mechanisms of resistance led to the use of alternative agents that improved clinical benefit, demonstrating that an understanding of the molecular mechanisms driving resistance to specific therapies is of paramount importance. Here we review the most generalized mechanisms of resistance to targeted therapies, together with some experimental strategies employed to identify such mechanisms. Therapeutic failure is not an option and we need to understand the dynamics of tumor adaptation in order to adequately adjust therapies; in essence 'to fight fire with fire'.
Collapse
|
1856
|
Cheng X, Chen H. Tumor heterogeneity and resistance to EGFR-targeted therapy in advanced nonsmall cell lung cancer: challenges and perspectives. Onco Targets Ther 2014; 7:1689-704. [PMID: 25285017 PMCID: PMC4181629 DOI: 10.2147/ott.s66502] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lung cancer, mostly nonsmall cell lung cancer, continues to be the leading cause of cancer-related death worldwide. With the development of tyrosine kinase inhibitors that selectively target lung cancer-related epidermal growth factor receptor mutations, management of advanced nonsmall cell lung cancer has been greatly transformed. Improvements in progression-free survival and life quality of the patients were observed in numerous clinical studies. However, overall survival is not prolonged because of later-acquired drug resistance. Recent studies reveal a heterogeneous subclonal architecture of lung cancer, so it is speculated that the tumor may rapidly adapt to environmental changes via a Darwinian selection mechanism. In this review, we aim to provide an overview of both spatial and temporal tumor heterogeneity as potential mechanisms underlying epidermal growth factor receptor tyrosine kinase inhibitor resistance in nonsmall cell lung cancer and summarize the possible origins of tumor heterogeneity covering theories of cancer stem cells and clonal evolution, as well as genomic instability and epigenetic aberrations in lung cancer. Moreover, investigational measures that overcome heterogeneity-associated drug resistance and new assays to improve tumor assessment are also discussed.
Collapse
Affiliation(s)
- Xinghua Cheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| |
Collapse
|
1857
|
Bodnar M, Szylberg Ł, Kazmierczak W, Marszalek A. Tumor progression driven by pathways activating matrix metalloproteinases and their inhibitors. J Oral Pathol Med 2014; 44:437-43. [PMID: 25244188 DOI: 10.1111/jop.12270] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Laryngeal squamous cell carcinoma (LSCC) is still a problem worldwide. In some publications interactions between the expression of matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, and their tissue inhibitors (TIMPs) implicated during cancer progression were suggested. METHODS The immunohistochemical staining using primary antibody against MMP-2, MMP-9, TIMP-1, TIMP-2, and TIMP-3 were performed. The research group consists of primary N(0) LSCC (20 cases), primary N(+) LSCC (17 cases), and 18 cases of normal mucosa. RESULTS Studied MMPs and TIMPs were localized in tumor cells and tumor stroma compartment. MMP-2 expression was higher in stroma compared to tumor cells. MMP-9, TIMP-1, TIMP-2, and TIMP-3 expression was higher in tumor cells than in tumor stroma (P < 0.05). In tumor stroma MMP-2, MMP-9, TIMP-1, and TIMP-3 expression, in LSCC N(0) vs. LSCC N(+) was significantly higher (P < 0.05). The ratios between MMP-2 and TIMP-3 expression were statistically significant (N(0) vs. N(+); P = 0.012). The analyses using classification trees predicted the probability of metastases according to TIMP-3/MMP-14/MMP-2 and MMP-9/TIMP-1 expression levels. CONCLUSIONS The presence of MMP-2, MMP-9, TIMP-1, TIMP-2, TIMP-3 expression in tumor cells and in tumor stroma, and additionally different expression according to lymph node involvement suggested of their impact during cancer progression. The significant correlation between TIMP-3 expression and the presence of lymph node metastases and MMP-2 expression might suggest the importance of TIMP-3 as a prognostic factor during tumor progression. The evaluation of molecular markers which participate in MMP-2 activation pathway have a major impact during metastasis.
Collapse
Affiliation(s)
- Magdalena Bodnar
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Wojciech Kazmierczak
- Department of Otolaryngology and Clinical Oncology Chair and Clinic of Otolaryngology and Department of Pathophysiology of Hearing and Balance System, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Andrzej Marszalek
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Bydgoszcz, Poland.,Department of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, Poland
| |
Collapse
|
1858
|
Elitas M, Brower K, Lu Y, Chen JJ, Fan R. A microchip platform for interrogating tumor-macrophage paracrine signaling at the single-cell level. LAB ON A CHIP 2014; 14:3582-8. [PMID: 25057779 PMCID: PMC4145007 DOI: 10.1039/c4lc00676c] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
It is increasingly recognized that infiltrating immune cells contribute to the pathogenesis of a wide range of solid tumors. The paracrine signaling between the tumor and the immune cells alters the functional state of individual tumor cells and, correspondingly, the anticipated response to radiation or chemotherapies, which is of great importance to clinical oncology. Here we present a high-density microchip platform capable of measuring a panel of paracrine signals associated with heterotypic tumor-immune cell interactions in the single-cell, pair-wise manner. The device features a high-content cell capture array of 5000+ sub-nanoliter microchambers for the isolation of single and multi-cell combinations and a multi-plex antibody "barcode" array for multiplexed protein secretion analysis from each microchamber. In this work, we measured a panel of 16 proteins produced from individual glioma cells, individual macrophage cells and varying heterotypic multi-cell combinations of both on the same device. The results show changes of tumor cell functional phenotypes that cannot be explained by an additive effect from isolated single cells and, presumably, can be attributed to the paracrine signaling between macrophage and glioma cells. The protein correlation analysis reveals the key signaling nodes altered by tumor-macrophage communication. This platform enables the novel pair-wise interrogation of heterotypic cell-cell paracrine signaling at the individual cell level with an in-depth analysis of the changing functional phenotypes for different co-culture cell combinations.
Collapse
Affiliation(s)
- Meltem Elitas
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520, USA.
| | | | | | | | | |
Collapse
|
1859
|
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
|
1860
|
Wang N, Gong T, Clarke R, Chen L, Shih IM, Zhang Z, Levine DA, Xuan J, Wang Y. UNDO: a Bioconductor R package for unsupervised deconvolution of mixed gene expressions in tumor samples. ACTA ACUST UNITED AC 2014; 31:137-9. [PMID: 25212756 DOI: 10.1093/bioinformatics/btu607] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SUMMARY We develop a novel unsupervised deconvolution method, within a well-grounded mathematical framework, to dissect mixed gene expressions in heterogeneous tumor samples. We implement an R package, UNsupervised DecOnvolution (UNDO), that can be used to automatically detect cell-specific marker genes (MGs) located on the scatter radii of mixed gene expressions, estimate cellular proportions in each sample and deconvolute mixed expressions into cell-specific expression profiles. We demonstrate the performance of UNDO over a wide range of tumor-stroma mixing proportions, validate UNDO on various biologically mixed benchmark gene expression datasets and further estimate tumor purity in TCGA/CPTAC datasets. The highly accurate deconvolution results obtained suggest not only the existence of cell-specific MGs but also UNDO's ability to detect them blindly and correctly. Although the principal application here involves microarray gene expressions, our methodology can be readily applied to other types of quantitative molecular profiling data. AVAILABILITY AND IMPLEMENTATION UNDO is available at http://bioconductor.org/packages.
Collapse
Affiliation(s)
- Niya Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Ting Gong
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Robert Clarke
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Lulu Chen
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Ie-Ming Shih
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Zhen Zhang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Douglas A Levine
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, Departments of Pathology and Oncology, Johns Hopkins University, Baltimore, MD 21231 and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| |
Collapse
|
1861
|
Rigoni A, Colombo MP, Pucillo C. The Role of Mast Cells in Molding the Tumor Microenvironment. CANCER MICROENVIRONMENT 2014; 8:167-76. [PMID: 25194694 DOI: 10.1007/s12307-014-0152-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 08/20/2014] [Indexed: 12/20/2022]
Abstract
Mast cells (MCs) are granulocytic immune cells that reside in tissues exposed to the external environment. MCs are best known for their activity in allergic reactions, but they have been involved in different physiological and pathological conditions. In particular, MC infiltration has been shown in several types of human tumors and in animal cancer models. Nevertheless, the role of MCs in the tumor microenvironment is still debated because they have been associated either to good or poor prognosis depending on tumor type and tissue localization. This dichotomous role relies on MC capacity to secrete a broad spectrum of molecules with modulatory functions, which may condition the final tumor outcome also promoting angiogenesis and tissue remodeling. In this review, we analyze the multifaceted role of mast cell in tumor progression and inhibition considering their ability to interact with: i) immune cells, ii) tumor cells and iii) the extracellular matrix. Eventually, the current MC targeting strategies to treat cancer patients are discussed. Deciphering the actual role of MCs in tumor onset and progression is crucial to identify MC-targeted treatments aimed at killing cancer cells or at making the tumor vulnerable to selected anti-cancer drugs.
Collapse
Affiliation(s)
- A Rigoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Amadeo 42, 20133, Milan, Italy
| | - M P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Amadeo 42, 20133, Milan, Italy.
| | - C Pucillo
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| |
Collapse
|
1862
|
Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, Clarke RB, de Jong S, Jonkers J, Mælandsmo GM, Roman-Roman S, Seoane J, Trusolino L, Villanueva A. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov 2014; 4:998-1013. [PMID: 25185190 PMCID: PMC4167608 DOI: 10.1158/2159-8290.cd-14-0001] [Citation(s) in RCA: 1239] [Impact Index Per Article: 112.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED Recently, there has been an increasing interest in the development and characterization of patient-derived tumor xenograft (PDX) models for cancer research. PDX models mostly retain the principal histologic and genetic characteristics of their donor tumor and remain stable across passages. These models have been shown to be predictive of clinical outcomes and are being used for preclinical drug evaluation, biomarker identification, biologic studies, and personalized medicine strategies. This article summarizes the current state of the art in this field, including methodologic issues, available collections, practical applications, challenges and shortcomings, and future directions, and introduces a European consortium of PDX models. SIGNIFICANCE PDX models are increasingly used in translational cancer research. These models are useful for drug screening, biomarker development, and the preclinical evaluation of personalized medicine strategies. This review provides a timely overview of the key characteristics of PDX models and a detailed discussion of future directions in the field.
Collapse
Affiliation(s)
| | | | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow
| | | | | | | | - Robert B Clarke
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Jos Jonkers
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | | | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS; and Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Alberto Villanueva
- Catalan Institute of Oncology-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
1863
|
Sempere LF. Tissue slide-based microRNA characterization of tumors: how detailed could diagnosis become for cancer medicine? Expert Rev Mol Diagn 2014; 14:853-69. [PMID: 25090088 PMCID: PMC4364265 DOI: 10.1586/14737159.2014.944507] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
miRNAs are short, non-coding, regulatory RNAs that exert cell type-dependent, context-dependent, transcriptome-wide gene expression control under physiological and pathological conditions. Tissue slide-based assays provide qualitative (tumor compartment) and semi-quantitative (expression levels) information about altered miRNA expression at single-cell resolution in clinical tumor specimens. Reviewed here are key technological advances in the last 5 years that have led to implementation of fully automated, robust and reproducible tissue slide-based assays for in situ miRNA detection on US FDA-approved instruments; recent tissue slide-based discovery studies that suggest potential clinical applications of specific miRNAs in cancer medicine are highlighted; and the challenges in bringing tissue slide-based miRNA assays into the clinic are discussed, including clinical validation, biomarker performance, biomarker space and integration with other biomarkers.
Collapse
Affiliation(s)
- Lorenzo F Sempere
- Laboratory of microRNA Diagnostics and Therapeutics, Van Andel Research Institute, 333 Bostwick Ave, N.E, Grand Rapids, MI 49503, USA
| |
Collapse
|
1864
|
Henderson D, Ogilvie LA, Hoyle N, Keilholz U, Lange B, Lehrach H. Personalized medicine approaches for colon cancer driven by genomics and systems biology: OncoTrack. Biotechnol J 2014; 9:1104-14. [PMID: 25074435 PMCID: PMC4314672 DOI: 10.1002/biot.201400109] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 05/20/2014] [Accepted: 06/26/2014] [Indexed: 12/15/2022]
Abstract
The post-genomic era promises to pave the way to a personalized understanding of disease processes, with technological and analytical advances helping to solve some of the world's health challenges. Despite extraordinary progress in our understanding of cancer pathogenesis, the disease remains one of the world's major medical problems. New therapies and diagnostic procedures to guide their clinical application are urgently required. OncoTrack, a consortium between industry and academia, supported by the Innovative Medicines Initiative, signifies a new era in personalized medicine, which synthesizes current technological advances in omics techniques, systems biology approaches, and mathematical modeling. A truly personalized molecular imprint of the tumor micro-environment and subsequent diagnostic and therapeutic insight is gained, with the ultimate goal of matching the "right" patient to the "right" drug and identifying predictive biomarkers for clinical application. This comprehensive mapping of the colon cancer molecular landscape in tandem with crucial, clinical functional annotation for systems biology analysis provides unprecedented insight and predictive power for colon cancer management. Overall, we show that major biotechnological developments in tandem with changes in clinical thinking have laid the foundations for the OncoTrack approach and the future clinical application of a truly personalized approach to colon cancer theranostics.
Collapse
|
1865
|
Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O’Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol 2014; 15:433. [PMID: 25159823 PMCID: PMC4166471 DOI: 10.1186/s13059-014-0433-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/08/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genomic analysis of multi-focal renal cell carcinomas from an individual with a germline VHL mutation offers a unique opportunity to study tumor evolution. RESULTS We perform whole exome sequencing on four clear cell renal cell carcinomas removed from both kidneys of a patient with a germline VHL mutation. We report that tumors arising in this context are clonally independent and harbour distinct secondary events exemplified by loss of chromosome 3p, despite an identical genetic background and tissue microenvironment. We propose that divergent mutational and copy number anomalies are contingent upon the nature of 3p loss of heterozygosity occurring early in tumorigenesis. However, despite distinct 3p events, genomic, proteomic and immunohistochemical analyses reveal evidence for convergence upon the PI3K-AKT-mTOR signaling pathway. Four germline tumors in this young patient, and in a second, older patient with VHL syndrome demonstrate minimal intra-tumor heterogeneity and mutational burden, and evaluable tumors appear to follow a linear evolutionary route, compared to tumors from patients with sporadic clear cell renal cell carcinoma. CONCLUSIONS In tumors developing from a germline VHL mutation, the evolutionary principles of contingency and convergence in tumor development are complementary. In this small set of patients with early stage VHL-associated tumors, there is reduced mutation burden and limited evidence of intra-tumor heterogeneity.
Collapse
Affiliation(s)
- Rosalie Fisher
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - Stuart Horswell
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Andrew Rowan
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | | | - Elza C de Bruin
- />University College London Cancer Institute, London, WC1E 6DD UK
| | - Sakshi Gulati
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Nicholas McGranahan
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
- />Centre for Mathematics & Physics in the Life Science & Experimental Biology (CoMPLEX), University College London, London, WC1E 6BT UK
| | - Mark Stares
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - Marco Gerlinger
- />Centre for Evolution and Cancer, Institute of Cancer Research, London, SW7 3RP UK
| | - Ignacio Varela
- />Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC-Sodercan), Departamento de Biología Molecular, Universidad de Cantabria, Santander, 39011 Spain
| | - Andrew Crockford
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Francesco Favero
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
- />Cancer System Biology, Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, DK-2800 Denmark
| | | | | | - Carolina Navas
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Eva Grönroos
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - David Nicol
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - Steve Hazell
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - David Hrouda
- />Imperial College Healthcare NHS Trust, London, W6 8RF UK
| | - Tim O’Brien
- />Guy’s and St Thomas’ NHS Foundation Trust, London, SE1 9RT UK
| | - Nik Matthews
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Ben Phillimore
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Sharmin Begum
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Adam Rabinowitz
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Jennifer Biggs
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Paul A Bates
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Neil Q McDonald
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | - Gordon Stamp
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
| | | | - James J Hsieh
- />Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, 10065 USA
| | - Jianing Xu
- />Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, 10065 USA
| | - Lisa Pickering
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - Martin Gore
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - James Larkin
- />Royal Marsden NHS Foundation Trust, London, SW3 6JJ UK
| | - Charles Swanton
- />Cancer Research UK London Research Institute, London, WC2A 3LY UK
- />University College London Cancer Institute, London, WC1E 6DD UK
| |
Collapse
|
1866
|
Pacheco JM, Santos FC, Dingli D. The ecology of cancer from an evolutionary game theory perspective. Interface Focus 2014; 4:20140019. [PMID: 25097748 DOI: 10.1098/rsfs.2014.0019] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The accumulation of somatic mutations, to which the cellular genome is permanently exposed, often leads to cancer. Analysis of any tumour shows that, besides the malignant cells, one finds other 'supporting' cells such as fibroblasts, immune cells of various types and even blood vessels. Together, these cells generate the microenvironment that enables the malignant cell population to grow and ultimately lead to disease. Therefore, understanding the dynamics of tumour growth and response to therapy is incomplete unless the interactions between the malignant cells and normal cells are investigated in the environment in which they take place. The complex interactions between cells in such an ecosystem result from the exchange of information in the form of cytokines- and adhesion-dependent interactions. Such processes impose costs and benefits to the participating cells that may be conveniently recast in the form of a game pay-off matrix. As a result, tumour progression and dynamics can be described in terms of evolutionary game theory (EGT), which provides a convenient framework in which to capture the frequency-dependent nature of ecosystem dynamics. Here, we provide a tutorial review of the central aspects of EGT, establishing a relation with the problem of cancer. Along the way, we also digress on fitness and of ways to compute it. Subsequently, we show how EGT can be applied to the study of the various manifestations and dynamics of multiple myeloma bone disease and its preceding condition known as monoclonal gammopathy of undetermined significance. We translate the complex biochemical signals into costs and benefits of different cell types, thus defining a game pay-off matrix. Then we use the well-known properties of the EGT equations to reduce the number of core parameters that characterize disease evolution. Finally, we provide an interpretation of these core parameters in terms of what their function is in the ecosystem we are describing and generate predictions on the type and timing of interventions that can alter the natural history of these two conditions.
Collapse
Affiliation(s)
- Jorge M Pacheco
- Departamento de Matemática e Aplicações , Universidade do Minho , Braga 4710-057 , Portugal ; Centro de Biologia Molecular e Ambiental , Universidade do Minho , Braga 4710-057 , Portugal ; ATP-Group, CMAF , Instituto para a Investigação Interdisciplinar , Lisboa 1649-003 , Portugal
| | - Francisco C Santos
- INESC-ID and Instituto Superior Técnico , Universidade de Lisboa , Taguspark, Porto Salvo, Lisboa 2744-016 , Portugal ; ATP-Group, CMAF , Instituto para a Investigação Interdisciplinar , Lisboa 1649-003 , Portugal
| | - David Dingli
- Division of Hematology and Department of Molecular Medicine , Mayo Clinic , 200 First Street SW, Rochester, MN 55905 , USA
| |
Collapse
|
1867
|
Anderson JL, Titz B, Akiyama R, Komisopoulou E, Park A, Tap WD, Graeber TG, Denny CT. Phosphoproteomic profiling reveals IL6-mediated paracrine signaling within the Ewing sarcoma family of tumors. Mol Cancer Res 2014; 12:1740-54. [PMID: 25092916 DOI: 10.1158/1541-7786.mcr-14-0159] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Members of the Ewing sarcoma family of tumors (ESFT) contain tumor-associated translocations that give rise to oncogenic transcription factors, most commonly EWS/FLI1. EWS/FLI1 plays a dominant role in tumor progression by modulating the expression of hundreds of target genes. Here, the impact of EWS/FLI1 inhibition, by RNAi-mediated knockdown, on cellular signaling was investigated using mass spectrometry-based phosphoproteomics to quantify global changes in phosphorylation. This unbiased approach identified hundreds of unique phosphopeptides enriched in processes such as regulation of cell cycle and cytoskeleton organization. In particular, phosphotyrosine profiling revealed a large upregulation of STAT3 phosphorylation upon EWS/FLI1 knockdown. However, single-cell analysis demonstrated that this was not a cell-autonomous effect of EWS/FLI1 deficiency, but rather a signaling effect occurring in cells in which knockdown does not occur. Conditioned media from knockdown cells were sufficient to induce STAT3 phosphorylation in control cells, verifying the presence of a soluble factor that can activate STAT3. Cytokine analysis and ligand/receptor inhibition experiments determined that this activation occurred, in part, through an IL6-dependent mechanism. Taken together, the data support a model in which EWS/FLI1 deficiency results in the secretion of soluble factors, such as IL6, which activate STAT signaling in bystander cells that maintain EWS/FLI1 expression. Furthermore, these soluble factors were shown to protect against apoptosis. IMPLICATIONS EWS/FLI1 inhibition results in a novel adaptive response and suggests that targeting the IL6/STAT3 signaling pathway may increase the efficacy of ESFT therapies.
Collapse
Affiliation(s)
- Jennifer L Anderson
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California. Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, University of California, Los Angeles, Los Angeles, California
| | - Björn Titz
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ryan Akiyama
- Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, University of California, Los Angeles, Los Angeles, California
| | - Evangelia Komisopoulou
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ann Park
- Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, University of California, Los Angeles, Los Angeles, California
| | - William D Tap
- Sarcoma Medical Oncology Service, Division of Solid Tumors, Department of Medicine, Memorial Sloan Kettering Cancer Center and Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Thomas G Graeber
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California. California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California. UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, California
| | - Christopher T Denny
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California. Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, University of California, Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California. California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
1868
|
Translation in solid cancer: are size-based response criteria an anachronism? Clin Transl Oncol 2014; 17:1-10. [PMID: 25073600 DOI: 10.1007/s12094-014-1207-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/09/2014] [Indexed: 12/19/2022]
Abstract
The purpose of translation is the development of effective medicinal products based on validated science. A parallel objective is to obtain marketing authorization for the translated product. Unfortunately, in solid cancer, these two objectives are not mutually consistent as evidenced by the contrast between major advances in science and the continuing dismal record of pharmaceutical productivity. If the problem is unrelated to science, then the process of translation may require a closer examination, namely, the criteria for regulatory approval. This realization is important because, in this context, the objective of translation is regulatory approval, and science does not passively translate into useful medicinal products. Today, in solid cancer, response criteria related to tumor size are less useful than during the earlier cytotoxic drugs era; advanced imaging and biomarkers now allow for tracking of the natural history of the disease in the laboratory and the clinic. Also, it is difficult to infer clinical benefit from tumor shrinkage since it is rarely sustained. Accordingly, size-based response criteria may represent an anachronism relative to translation in solid cancer and it may be appropriate to align preclinical and clinical effort and shift the focus to local invasion and metastasis. The shift from a cancer cell-centric model to a stroma centric model offers novel opportunities not only to interupt the natural history of the disease, but also to rethink the relevance of outdated criteria of clinical response. Current evidence favors the opinion that, in solid cancer, a different, broader, and contextual approach may lead to interventions that could delay local invasion and metastasis. All elements supporting this shift, especially advanced imaging, are in place.
Collapse
|
1869
|
Using real-time impedance-based assays to monitor the effects of fibroblast-derived media on the adhesion, proliferation, migration and invasion of colon cancer cells. Biosci Rep 2014; 34:BSR20140031. [PMID: 24935351 PMCID: PMC4114067 DOI: 10.1042/bsr20140031] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Increasing our knowledge of the mechanisms regulating cell proliferation, migration and invasion are central to understanding tumour progression and metastasis. The local tumour microenvironment contributes to the transformed phenotype in cancer by providing specific environmental cues that alter the cells behaviour and promotes metastasis. Fibroblasts have a strong association with cancer and in recent times there has been some emphasis in designing novel therapeutic strategies that alter fibroblast behaviour in the tumour microenvironment. Fibroblasts produce growth factors, chemokines and many of the proteins laid down in the ECM (extracellular matrix) that promote angiogenesis, inflammation and tumour progression. In this study, we use a label-free RTCA (real-time cell analysis) platform (xCELLigence) to investigate how media derived from human fibroblasts alters cancer cell behaviour. We used a series of complimentary and novel experimental approaches to show HCT116 cells adhere, proliferate and migrate significantly faster in the presence of media from human fibroblasts. As well as this, we used the xCELLigence CIM-plates system to show that HCT116 cells invade matrigel layers aggressively when migrating towards media derived from human fibroblasts. These data strongly suggest that fibroblasts have the ability to increase the migratory and invasive properties of HCT116 cells. This is the first study that provides real-time data on fibroblast-mediated migration and invasion kinetics of colon cancer cells.
Collapse
|
1870
|
Sriraman SK, Aryasomayajula B, Torchilin VP. Barriers to drug delivery in solid tumors. Tissue Barriers 2014; 2:e29528. [PMID: 25068098 PMCID: PMC4106925 DOI: 10.4161/tisb.29528] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/06/2014] [Accepted: 06/09/2014] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, significant progress has been made in the field of drug delivery. The advent of engineered nanoparticles has allowed us to circumvent the initial limitations to drug delivery such as pharmacokinetics and solubility. However, in spite of significant advances to tumor targeting, an effective treatment strategy for malignant tumors still remains elusive. Tumors possess distinct physiological features which allow them to resist traditional treatment approaches. This combined with the complexity of the biological system presents significant hurdles to the site-specific delivery of therapeutic drugs. One of the key features of engineered nanoparticles is that these can be tailored to execute specific functions. With this review, we hope to provide the reader with a clear understanding and knowledge of biological barriers and the methods to exploit these characteristics to design multifunctional nanocarriers, effect useful dosing regimens and subsequently improve therapeutic outcomes in the clinic.
Collapse
Affiliation(s)
- Shravan Kumar Sriraman
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| | - Bhawani Aryasomayajula
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| |
Collapse
|
1871
|
Gough AH, Chen N, Shun TY, Lezon TR, Boltz RC, Reese CE, Wagner J, Vernetti LA, Grandis JR, Lee AV, Stern AM, Schurdak ME, Taylor DL. Identifying and quantifying heterogeneity in high content analysis: application of heterogeneity indices to drug discovery. PLoS One 2014; 9:e102678. [PMID: 25036749 PMCID: PMC4103836 DOI: 10.1371/journal.pone.0102678] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/22/2014] [Indexed: 12/04/2022] Open
Abstract
One of the greatest challenges in biomedical research, drug discovery and diagnostics is understanding how seemingly identical cells can respond differently to perturbagens including drugs for disease treatment. Although heterogeneity has become an accepted characteristic of a population of cells, in drug discovery it is not routinely evaluated or reported. The standard practice for cell-based, high content assays has been to assume a normal distribution and to report a well-to-well average value with a standard deviation. To address this important issue we sought to define a method that could be readily implemented to identify, quantify and characterize heterogeneity in cellular and small organism assays to guide decisions during drug discovery and experimental cell/tissue profiling. Our study revealed that heterogeneity can be effectively identified and quantified with three indices that indicate diversity, non-normality and percent outliers. The indices were evaluated using the induction and inhibition of STAT3 activation in five cell lines where the systems response including sample preparation and instrument performance were well characterized and controlled. These heterogeneity indices provide a standardized method that can easily be integrated into small and large scale screening or profiling projects to guide interpretation of the biology, as well as the development of therapeutics and diagnostics. Understanding the heterogeneity in the response to perturbagens will become a critical factor in designing strategies for the development of therapeutics including targeted polypharmacology.
Collapse
Affiliation(s)
- Albert H. Gough
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Ning Chen
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Tong Ying Shun
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Timothy R. Lezon
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Robert C. Boltz
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Celeste E. Reese
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jacob Wagner
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lawrence A. Vernetti
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jennifer R. Grandis
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Adrian V. Lee
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Andrew M. Stern
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mark E. Schurdak
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - D. Lansing Taylor
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
1872
|
Zhu J, Huang H, Dong S, Ge L, Zhang Y. Progress in aptamer-mediated drug delivery vehicles for cancer targeting and its implications in addressing chemotherapeutic challenges. Theranostics 2014; 4:931-44. [PMID: 25057317 PMCID: PMC4107293 DOI: 10.7150/thno.9663] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/23/2014] [Indexed: 12/28/2022] Open
Abstract
Aptamers are novel oligonucleotides with flexible three-dimensional configurations that recognize and bind to their cognate targets, including tumor surface receptors, in a high-affinity and highly specific manner. Because of their unique intrinsic properties, a variety of aptamer-mediated nanovehicles have been developed to directionally transport anti-cancer drugs to tumor sites to minimize systemic cytotoxicity and to enhance permeation by these tumoricidal agents. Despite advances in the selection and synthesis of aptamers and in the conjugation and self-assembly of nanotechnologies, current chemotherapy and drug delivery systems face great challenges. These challenges are due to the limitations of aptamers and vehicles and because of complicated tumor mechanisms, including heterogeneity, anti-cancer drug resistance, and hypoxia-induced aberrances. In this review, we will summarize current approaches utilizing tumor surface hallmarks and aptamers and their roles and mechanisms in therapeutic nanovehicles targeting tumors. Delivery forms include nanoparticles, nanotubes, nanogels, aptamer-drug conjugates, and novel molecular trains. Moreover, the obstacles posed by the aforementioned issues will be highlighted, and possible solutions will be acknowledged. Furthermore, future perspectives will be presented, including cutting-edge integration with RNA interference nanotechnology and personalized chemotherapy, which will facilitate innovative approaches to aptamer-based therapeutics.
Collapse
|
1873
|
Ghesquière B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature 2014; 511:167-76. [DOI: 10.1038/nature13312] [Citation(s) in RCA: 350] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/08/2014] [Indexed: 12/11/2022]
|
1874
|
Fernández R, Lage S, Abad-García B, Barceló-Coblijn G, Terés S, López DH, Guardiola-Serrano F, Martín ML, Escribá PV, Fernández JA. Analysis of the lipidome of xenografts using MALDI-IMS and UHPLC-ESI-QTOF. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2014; 25:1237-1246. [PMID: 24760294 DOI: 10.1007/s13361-014-0882-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/10/2014] [Accepted: 03/10/2014] [Indexed: 06/03/2023]
Abstract
Human tumor xenografts in immunodeficient mice are a very popular model to study the development of cancer and to test new drug candidates. Among the parameters analyzed are the variations in the lipid composition, as they are good indicators of changes in the cellular metabolism. Here, we present a study on the distribution of lipids in xenografts of NCI-H1975 human lung cancer cells, using MALDI imaging mass spectrometry and UHPLC-ESI-QTOF. The identification of lipids directly from the tissue by MALDI was aided by the comparison with identification using ESI ionization in lipid extracts from the same xenografts. Lipids belonging to PCs, PIs, SMs, DAG, TAG, PS, PA, and PG classes were identified and their distribution over the xenograft was determined. Three areas were identified in the xenograft, corresponding to cells in different metabolic stages and to a layer of adipose tissue that covers the xenograft.
Collapse
Affiliation(s)
- Roberto Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1875
|
Insights into the role of components of the tumor microenvironment in oral carcinoma call for new therapeutic approaches. Exp Cell Res 2014; 325:58-64. [DOI: 10.1016/j.yexcr.2013.12.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 12/28/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022]
|
1876
|
Abstract
Cells within tumours have diverse genomes and epigenomes and interact differentially with their surrounding microenvironment generating intratumour heterogeneity, which has critical implications for treating cancer patients. Understanding the cellular and microenvironment composition and characteristics in individual tumours is critical to stratify the patient population that is likely to benefit from specific treatment regimens. Here, we will review the current understanding of intratumour heterogeneity at the genomic, epigenomic and microenvironmental levels. We will also discuss the clinical implications and the challenges posed by intratumour heterogeneity and evaluate noninvasive methods such as circulating biomarkers to characterize the cellular diversity of tumours. Comprehensive assessment of the molecular features of patients based on tumour specimen characterization (including intratumour spatial and temporal variations), ancillary noninvasive methods (such as circulating biomarkers and molecular imaging approaches) and the correct design of clinical trials are required to guide administration of targeted therapy and to control therapeutic resistance. Finding the means to accurately determine and effectively control tumour heterogeneity and translate these achievements into patient benefit are major goals in modern oncology.
Collapse
Affiliation(s)
- J Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | |
Collapse
|
1877
|
HER2/neu: an increasingly important therapeutic target. Part 1: basic biology & therapeutic armamentarium. ACTA ACUST UNITED AC 2014. [DOI: 10.4155/cli.14.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
1878
|
Stebbing J, Paz K, Schwartz GK, Wexler LH, Maki R, Pollock RE, Morris R, Cohen R, Shankar A, Blackman G, Harding V, Vasquez D, Krell J, Ciznadija D, Katz A, Sidransky D. Patient-derived xenografts for individualized care in advanced sarcoma. Cancer 2014; 120:2006-15. [PMID: 24705963 PMCID: PMC4298787 DOI: 10.1002/cncr.28696] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/18/2013] [Accepted: 01/02/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Patients with advanced, metastatic sarcoma have a poor prognosis, and the overall benefit from the few standard-of-care therapeutics available is small. The rarity of this tumor, combined with the wide range of subtypes, leads to difficulties in conducting clinical trials. The authors previously reported the outcome of patients with a variety of common solid tumors who received treatment with drug regimens that were first tested in patient-derived xenografts using a proprietary method ("TumorGrafts"). METHODS Tumors resected from 29 patients with sarcoma were implanted into immunodeficient mice to identify drug targets and drugs for clinical use. The results of drug sensitivity testing in the TumorGrafts were used to personalize cancer treatment. RESULTS Of 29 implanted tumors, 22 (76%) successfully engrafted, permitting the identification of treatment regimens for these patients. Although 6 patients died before the completion of TumorGraft testing, a correlation between TumorGraft results and clinical outcome was observed in 13 of 16 (81%) of the remaining individuals. No patients progressed during the TumorGraft-predicted therapy. CONCLUSIONS The current data support the use of the personalized TumorGraft model as an investigational platform for therapeutic decision-making that can guide treatment for rare tumors such as sarcomas. A randomized phase 3 trial versus physician's choice is warranted.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of Oncology, Imperial College and Imperial Healthcare National Health Service Trust, Hammersmith HospitalLondon, United Kingdom
| | - Keren Paz
- Department of Oncology, Champions OncologyBaltimore, Maryland
| | - Gary K Schwartz
- Department of Oncology, Memorial Sloan-Kettering Cancer CenterNew York, New York
| | - Leonard H Wexler
- Department of Oncology, Memorial Sloan-Kettering Cancer CenterNew York, New York
| | - Robert Maki
- Department of Oncology, Mount Sinai School of MedicineNew York, New York
| | - Raphael E Pollock
- Department of Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Ronnie Morris
- Department of Oncology, Champions OncologyBaltimore, Maryland
| | - Richard Cohen
- Department of Surgery, University College HospitalsLondon, United Kingdom
| | - Arjun Shankar
- Department of Surgery, University College HospitalsLondon, United Kingdom
| | - Glen Blackman
- Department of Radiotherapy, University College HospitalsLondon, United Kingdom
| | - Victoria Harding
- Department of Oncology, Imperial College and Imperial Healthcare National Health Service Trust, Hammersmith HospitalLondon, United Kingdom
| | - David Vasquez
- Department of Oncology, Champions OncologyBaltimore, Maryland
| | - Jonathan Krell
- Department of Oncology, Imperial College and Imperial Healthcare National Health Service Trust, Hammersmith HospitalLondon, United Kingdom
| | | | - Amanda Katz
- Department of Oncology, Champions OncologyBaltimore, Maryland
| | - David Sidransky
- Department of Oncology, Johns Hopkins University School of MedicineBaltimore, Maryland
| |
Collapse
|
1879
|
Abouelmagd SA, Hyun H, Yeo Y. Extracellularly activatable nanocarriers for drug delivery to tumors. Expert Opin Drug Deliv 2014; 11:1601-1618. [PMID: 24950343 DOI: 10.1517/17425247.2014.930434] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Nanoparticles (NPs) for drug delivery to tumors need to satisfy two seemingly conflicting requirements: they should maintain physical and chemical stability during circulation and be able to interact with target cells and release the drug at desired locations with no substantial delay. The unique microenvironment of tumors and externally applied stimuli provide a useful means to maintain a balance between the two requirements. AREAS COVERED We discuss nanoparticulate drug carriers that maintain stable structures in normal conditions but respond to stimuli for the spatiotemporal control of drug delivery. We first define the desired effects of extracellular activation of NPs and frequently used stimuli and then review the examples of extracellularly activated NPs. EXPERT OPINION Several challenges remain in developing extracellularly activatable NPs. First, some of the stimuli-responsive NPs undergo incremental changes in response to stimuli, losing circulation stability. Second, the applicability of stimuli in clinical settings is limited due to the occasional occurrence of the activating conditions in normal tissues. Third, the construction of stimuli-responsive NPs involves increasing complexity in NP structure and production methods. Future efforts are needed to identify new targeting conditions and increase the contrast between activated and nonactivated NPs while keeping the production methods simple and scalable.
Collapse
Affiliation(s)
- Sara A Abouelmagd
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Hyesun Hyun
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.,Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, USA
| |
Collapse
|
1880
|
Seth A, Heo MB, Lim YT. Poly (γ-glutamic acid) based combination of water-insoluble paclitaxel and TLR7 agonist for chemo-immunotherapy. Biomaterials 2014; 35:7992-8001. [PMID: 24954733 DOI: 10.1016/j.biomaterials.2014.05.076] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 05/26/2014] [Indexed: 12/21/2022]
Abstract
Advanced anti-cancer regimens are being introduced for more effective cancer treatment with improved life expectancy. In this research, immuno-stimulating agent toll-like receptor-7 (TLR-7) agonist-imiquimod and low dose chemotherapeutic agent-paclitaxel were synergized to demonstrate tumor therapy along with anti-tumor memory effect. Both therapeutic agents being water insoluble were dispersed in water with the help of water soluble polymer: poly (γ-glutamic acid) (γ-PGA) using a co-solvent systems leading to formation of micro-dispersions of drugs. Paclitaxel and imiquimod formed crystalline microstructures in the size range of 2-3 μm and were stably dispersed in γ-PGA matrix for more than 6 months. Paclitaxel and combination of paclitaxel and imiquimod had significant tumor killing effect in-vitro on various tumor cell lines, while antigen presenting cells (dendritic cells-DCs) treated with the same concentration of imiquimod along with the combination led to enhanced proliferation (250%). In DCs, enhanced secretion of pro-inflammatory and Th1 cytokines was observed in cells co-treated with paclitaxel and imiquimod dispersed in γ-PGA. When administered by intra-tumoral injection in mouse melanoma tumor model, the treatment with combination exemplified drastic inhibition of tumor growth leading to 70% survival as compared to individual components with 0% survival at day 41. The anti-tumor response generated was also found to have systemic memory response since the vaccinated mice significantly deferred secondary tumor development at distant site 6 weeks after treatment. The relative number and activation status of DCs in-vivo was found to be dramatically increased in case of mice treated with combination. The dramatic inhibition of tumor treated with combination is expected to be mediated by both chemotherapeutic killing of tumor cells followed by uptake of released antigen by the DCs and due to enhanced proliferation and activation of the DCs.
Collapse
Affiliation(s)
- Anushree Seth
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Min Beom Heo
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), School of Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
1881
|
Chicaybam L, Bonamino MH. Moving Receptor Redirected Adoptive Cell Therapy Toward Fine Tuning of Antitumor Responses. Int Rev Immunol 2014; 33:402-16. [DOI: 10.3109/08830185.2014.917412] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
1882
|
Dittmer J, Leyh B. The impact of tumor stroma on drug response in breast cancer. Semin Cancer Biol 2014; 31:3-15. [PMID: 24912116 DOI: 10.1016/j.semcancer.2014.05.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
In the last two decades the breast cancer mortality rate has steadily declined, in part, due to the availability of better treatment options. However, drug resistance still remains a major challenge. Resistance can be an inherent feature of breast cancer cells, but can also arise from the tumor microenvironment. This review aims to focus on the modulatory effect of the tumor microenvironment on the differing response of breast cancer subtypes to targeted drugs and chemotherapy.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany.
| | - Benjamin Leyh
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany
| |
Collapse
|
1883
|
Coulouarn C, Clément B. Stellate cells and the development of liver cancer: therapeutic potential of targeting the stroma. J Hepatol 2014; 60:1306-1309. [PMID: 24530649 DOI: 10.1016/j.jhep.2014.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are the most common types of primary tumors in the liver. Although major advances have been made in understanding the cellular and molecular mechanisms underlying liver carcinogenesis, HCC and ICC are still deadly cancers worldwide waiting for innovative therapeutic options. Growing evidence from the literature highlight the critical role of the tumor cell microenvironment in the pathogenesis of cancer diseases. Thus, targeting the microenvironment, particularly the crosstalk between tumor cells and stromal cells, has emerged as a promising therapeutic strategy. This strategy would be particularly relevant for liver cancers which frequently develop in a setting of chronic inflammation and microenvironment remodeling associated with hepatic fibrosis and cirrhosis, such processes in which hepatic stellate cells (HSC) greatly contribute. This review brings a genomic point of view on the alterations of the cellular microenvironment in liver cancers, particularly the stromal tissue within tumor nodules, emphasizing the importance of the crosstalk between tumor cells and stromal cells, notably activated HSC, in tumor onset and progression. Furthermore, potential therapeutic modalities of targeting the stroma and HSC are discussed.
Collapse
Affiliation(s)
- Cédric Coulouarn
- Inserm, UMR991, Liver Metabolisms and Cancer, F-35033 Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Bruno Clément
- Inserm, UMR991, Liver Metabolisms and Cancer, F-35033 Rennes, France; Université de Rennes 1, F-35043 Rennes, France
| |
Collapse
|
1884
|
Clark AM, Wheeler SE, Taylor DP, Pillai VC, Young CL, Prantil-Baun R, Nguyen T, Stolz DB, Borenstein JT, Lauffenburger DA, Venkataramanan R, Griffith LG, Wells A. A microphysiological system model of therapy for liver micrometastases. Exp Biol Med (Maywood) 2014; 239:1170-9. [PMID: 24821820 DOI: 10.1177/1535370214532596] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Metastasis accounts for almost 90% of cancer-associated mortality. The effectiveness of cancer therapeutics is limited by the protective microenvironment of the metastatic niche and consequently these disseminated tumors remain incurable. Metastatic disease progression continues to be poorly understood due to the lack of appropriate model systems. To address this gap in understanding, we propose an all-human microphysiological system that facilitates the investigation of cancer behavior in the liver metastatic niche. This existing LiverChip is a 3D-system modeling the hepatic niche; it incorporates a full complement of human parenchymal and non-parenchymal cells and effectively recapitulates micrometastases. Moreover, this system allows real-time monitoring of micrometastasis and assessment of human-specific signaling. It is being utilized to further our understanding of the efficacy of chemotherapeutics by examining the activity of established and novel agents on micrometastases under conditions replicating diurnal variations in hormones, nutrients and mild inflammatory states using programmable microdispensers. These inputs affect the cues that govern tumor cell responses. Three critical signaling groups are targeted: the glucose/insulin responses, the stress hormone cortisol and the gut microbiome in relation to inflammatory cues. Currently, the system sustains functioning hepatocytes for a minimum of 15 days; confirmed by monitoring hepatic function (urea, α-1-antitrypsin, fibrinogen, and cytochrome P450) and injury (AST and ALT). Breast cancer cell lines effectively integrate into the hepatic niche without detectable disruption to tissue, and preliminary evidence suggests growth attenuation amongst a subpopulation of breast cancer cells. xMAP technology combined with systems biology modeling are also employed to evaluate cellular crosstalk and illustrate communication networks in the early microenvironment of micrometastases. This model is anticipated to identify new therapeutic strategies for metastasis by elucidating the paracrine effects between the hepatic and metastatic cells, while concurrently evaluating agent efficacy for metastasis, metabolism and tolerability.
Collapse
Affiliation(s)
- Amanda M Clark
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Sarah E Wheeler
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Donald P Taylor
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Venkateswaran C Pillai
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Carissa L Young
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | | | - Transon Nguyen
- Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| | - Donna B Stolz
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | | | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | - Raman Venkataramanan
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | - Alan Wells
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| |
Collapse
|
1885
|
Yano S, Zhang Y, Miwa S, Tome Y, Hiroshima Y, Uehara F, Yamamoto M, Suetsugu A, Kishimoto H, Tazawa H, Zhao M, Bouvet M, Fujiwara T, Hoffman RM. Spatial-temporal FUCCI imaging of each cell in a tumor demonstrates locational dependence of cell cycle dynamics and chemoresponsiveness. Cell Cycle 2014; 13:2110-9. [PMID: 24811200 DOI: 10.4161/cc.29156] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The phase of the cell cycle can determine whether a cancer cell can respond to a given drug. We report here on the results of monitoring of real-time cell cycle dynamics of cancer cells throughout a live tumor intravitally using a fluorescence ubiquitination cell cycle indicator (FUCCI) before, during, and after chemotherapy. In nascent tumors in nude mice, approximately 30% of the cells in the center of the tumor are in G₀/G₁ and 70% in S/G₂/M. In contrast, approximately 90% of cancer cells in the center and 80% of total cells of an established tumor are in G₀/G₁ phase. Similarly, approximately 75% of cancer cells far from (> 100 µm) tumor blood vessels of an established tumor are in G₀/G₁. Longitudinal real-time imaging demonstrated that cytotoxic agents killed only proliferating cancer cells at the surface and, in contrast, had little effect on quiescent cancer cells, which are the vast majority of an established tumor. Moreover, resistant quiescent cancer cells restarted cycling after the cessation of chemotherapy. Our results suggest why most drugs currently in clinical use, which target cancer cells in S/G₂/M, are mostly ineffective on solid tumors. The results also suggest that drugs that target quiescent cancer cells are urgently needed.
Collapse
Affiliation(s)
- Shuya Yano
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA; Department of Gastroenterological Surgery; Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences; Okayama, Japan
| | | | - Shinji Miwa
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Yasunori Tome
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Yukihiko Hiroshima
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Fuminari Uehara
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Mako Yamamoto
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Atsushi Suetsugu
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Hiroyuki Kishimoto
- Department of Gastroenterological Surgery; Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences; Okayama, Japan
| | - Hiroshi Tazawa
- Center for Innovative Clinical Medicine; Okayama University Hospital; Okayama, Japan
| | | | - Michael Bouvet
- Department of Surgery; University of California, San Diego; La Jolla, CA USA
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery; Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences; Okayama, Japan
| | - Robert M Hoffman
- AntiCancer, Inc; San Diego, CA USA; Department of Surgery; University of California, San Diego; La Jolla, CA USA
| |
Collapse
|
1886
|
Zhao Y, Yao R, Ouyang L, Ding H, Zhang T, Zhang K, Cheng S, Sun W. Three-dimensional printing of Hela cells for cervical tumor model in vitro. Biofabrication 2014; 6:035001. [PMID: 24722236 DOI: 10.1088/1758-5082/6/3/035001] [Citation(s) in RCA: 320] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advances in three-dimensional (3D) printing have enabled the direct assembly of cells and extracellular matrix materials to form in vitro cellular models for 3D biology, the study of disease pathogenesis and new drug discovery. In this study, we report a method of 3D printing for Hela cells and gelatin/alginate/fibrinogen hydrogels to construct in vitro cervical tumor models. Cell proliferation, matrix metalloproteinase (MMP) protein expression and chemoresistance were measured in the printed 3D cervical tumor models and compared with conventional 2D planar culture models. Over 90% cell viability was observed using the defined printing process. Comparisons of 3D and 2D results revealed that Hela cells showed a higher proliferation rate in the printed 3D environment and tended to form cellular spheroids, but formed monolayer cell sheets in 2D culture. Hela cells in 3D printed models also showed higher MMP protein expression and higher chemoresistance than those in 2D culture. These new biological characteristics from the printed 3D tumor models in vitro as well as the novel 3D cell printing technology may help the evolution of 3D cancer study.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
1887
|
Shu Q, Li W, Li H, Sun G. Vasostatin inhibits VEGF-induced endothelial cell proliferation, tube formation and induces cell apoptosis under oxygen deprivation. Int J Mol Sci 2014; 15:6019-30. [PMID: 24722573 PMCID: PMC4013612 DOI: 10.3390/ijms15046019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/06/2014] [Accepted: 03/13/2014] [Indexed: 01/08/2023] Open
Abstract
Anti-angiogenesis treatment has been a promising new form of cancer therapy. Endothelial cells are critical for vascular homeostasis and play important roles in angiogenesis, vascular and tissue remodeling. Vasostatin, the 180 amino acid N-terminal fragment of the calreticulin protein, is reported to be a potent endogenous inhibitor of angiogenesis, suppressing tumor growth. However, the mechanism of these effects has not been sufficiently investigated. This study was performed to investigate the possible mechanism of vasostatin effects on primary cultured human umbilical vein endothelial cells (HUVEC). We found that vasostatin could inhibit the cell viability of HUVEC and induce cell apoptosis through mitochondrial pathways via activation of caspase-3 under oxygen deprivation conditions. Meanwhile, vasostatin also inhibited vascular endothelial growth factor-induced proliferation and tube formation of HUVEC. The possible mechanism of vasostatin-inhibited proliferation of HUVEC could be through down-regulation of endothelial nitric oxide synthase. These findings suggest that vasostatin could regulate endothelial cell function and might be used in anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Qun Shu
- Shanghai Changning Maternity and Infant Health Hospital, 773 Wuyi Road, Shanghai 200051, China.
| | - Wenjiao Li
- Shanghai Changning Maternity and Infant Health Hospital, 773 Wuyi Road, Shanghai 200051, China.
| | - Haichuan Li
- Shanghai Changning Maternity and Infant Health Hospital, 773 Wuyi Road, Shanghai 200051, China.
| | - Gang Sun
- School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, China.
| |
Collapse
|
1888
|
Thoma CR, Zimmermann M, Agarkova I, Kelm JM, Krek W. 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv Drug Deliv Rev 2014; 69-70:29-41. [PMID: 24636868 DOI: 10.1016/j.addr.2014.03.001] [Citation(s) in RCA: 335] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/19/2014] [Accepted: 03/05/2014] [Indexed: 12/17/2022]
Abstract
Phenotypic heterogeneity of cancer cells, cell biological context, heterotypic crosstalk and the microenvironment are key determinants of the multistep process of tumor development. They sign responsible, to a significant extent, for the limited response and resistance of cancer cells to molecular-targeted therapies. Better functional knowledge of the complex intra- and intercellular signaling circuits underlying communication between the different cell types populating a tumor tissue and of the systemic and local factors that shape the tumor microenvironment is therefore imperative. Sophisticated 3D multicellular tumor spheroid (MCTS) systems provide an emerging tool to model the phenotypic and cellular heterogeneity as well as microenvironmental aspects of in vivo tumor growth. In this review we discuss the cellular, chemical and physical factors contributing to zonation and cellular crosstalk within tumor masses. On this basis, we further describe 3D cell culture technologies for growth of MCTS as advanced tools for exploring molecular tumor growth determinants and facilitating drug discovery efforts. We conclude with a synopsis on technological aspects for on-line analysis and post-processing of 3D MCTS models.
Collapse
|
1889
|
Labarge MA, Parvin B, Lorens JB. Molecular deconstruction, detection, and computational prediction of microenvironment-modulated cellular responses to cancer therapeutics. Adv Drug Deliv Rev 2014; 69-70:123-31. [PMID: 24582543 DOI: 10.1016/j.addr.2014.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 02/14/2014] [Accepted: 02/18/2014] [Indexed: 12/21/2022]
Abstract
The field of bioengineering has pioneered the application of new precision fabrication technologies to model the different geometric, physical or molecular components of tissue microenvironments on solid-state substrata. Tissue engineering approaches building on these advances are used to assemble multicellular mimetic-tissues where cells reside within defined spatial contexts. The functional responses of cells in fabricated microenvironments have revealed a rich interplay between the genome and extracellular effectors in determining cellular phenotypes and in a number of cases have revealed the dominance of microenvironment over genotype. Precision bioengineered substrata are limited to a few aspects, whereas cell/tissue-derived microenvironments have many undefined components. Thus, introducing a computational module may serve to integrate these types of platforms to create reasonable models of drug responses in human tissues. This review discusses how combinatorial microenvironment microarrays and other biomimetic microenvironments have revealed emergent properties of cells in particular microenvironmental contexts, the platforms that can measure phenotypic changes within those contexts, and the computational tools that can unify the microenvironment-imposed functional phenotypes with underlying constellations of proteins and genes. Ultimately we propose that a merger of these technologies will enable more accurate pre-clinical drug discovery.
Collapse
Affiliation(s)
- Mark A Labarge
- Life Science Division of Lawrence Berkeley National Laboratory, University of California, 1 Cylcotron Rd, MS977, Berkeley, CA 94720, USA.
| | - Bahram Parvin
- Life Science Division of Lawrence Berkeley National Laboratory, University of California, 1 Cylcotron Rd, MS977, Berkeley, CA 94720, USA.
| | - James B Lorens
- Department of Biomedicine and Center for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5020 Bergen, Norway.
| |
Collapse
|
1890
|
Dotto GP. Multifocal epithelial tumors and field cancerization: stroma as a primary determinant. J Clin Invest 2014; 124:1446-53. [PMID: 24691479 DOI: 10.1172/jci72589] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It is increasingly evident that cancer results from altered organ homeostasis rather than from deregulated control of single cells or groups of cells. This applies especially to epithelial cancer, the most common form of human solid tumors and a major cause of cancer lethality. In the vast majority of cases, in situ epithelial cancer lesions do not progress into malignancy, even if they harbor many of the genetic changes found in invasive and metastatic tumors. While changes in tumor stroma are frequently viewed as secondary to changes in the epithelium, recent evidence indicates that they can play a primary role in both cancer progression and initiation. These processes may explain the phenomenon of field cancerization, i.e., the occurrence of multifocal and recurrent epithelial tumors that are preceded by and associated with widespread changes of surrounding tissue or organ "fields."
Collapse
|
1891
|
Schwitalla S. Tumor cell plasticity: the challenge to catch a moving target. J Gastroenterol 2014; 49:618-27. [PMID: 24566894 DOI: 10.1007/s00535-014-0943-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/04/2014] [Indexed: 02/04/2023]
Abstract
Every cancer cell is "different"--within one and the same tumor, between different lesions originating from the same tumor, among different patients suffering from the same tumor type, and certainly between different tumor types. The complexity of tumor development, with its genetic, phenotypic and functional heterogeneity and plasticity within tumors and between primary tumors and metastases, underlies the unpredictable influences and stimuli of a tumor-associated inflammatory microenvironment, immune response, mechanical and metabolic stress, therapy-induced inflammation or interaction with microbiota. The stochastic and context dependent nature of these factors accounts for the difficulties to investigate the impact of resulting cell plasticity on tumor development, and justifies the challenge to prevent tumor recurrence. The emerging concept of cell plasticity and reciprocity (to change the phenotype by processing signals from the environment) throws more light on the actual complexity of tumor heterogeneity than can be expected solely from a unidirectional, classical cancer stem cell (CSC) model. To date, it remains widely unclear to what extent cell plasticity impacts tumor development, and it is difficult to assess by current methods. As a high tumor plasticity is likely to predict a poor outcome for patients, the future therapeutic challenge will be the development of personalized treatment strategies to predict and finally prevent cell plasticity in patients.
Collapse
Affiliation(s)
- Sarah Schwitalla
- Dr. George Daley Laboratory, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, One Blackfan Circle, Boston, MA, 02115, USA,
| |
Collapse
|
1892
|
Combined cytolytic effects of a vaccinia virus encoding a single chain trimer of MHC-I with a Tax-epitope and Tax-specific CTLs on HTLV-I-infected cells in a rat model. BIOMED RESEARCH INTERNATIONAL 2014; 2014:902478. [PMID: 24791004 PMCID: PMC3985193 DOI: 10.1155/2014/902478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/20/2014] [Indexed: 02/01/2023]
Abstract
Adult T cell leukemia (ATL) is a malignant lymphoproliferative disease caused by human T cell leukemia virus type I (HTLV-I). To develop an effective therapy against the disease, we have examined the oncolytic ability of an attenuated vaccinia virus (VV), LC16m8Δ (m8Δ), and an HTLV-I Tax-specific cytotoxic T lymphocyte (CTL) line, 4O1/C8, against an HTLV-I-infected rat T cell line, FPM1. Our results demonstrated that m8Δ was able to replicate in and lyse tumorigenic FPM1 cells but was incompetent to injure 4O1/C8 cells, suggesting the preferential cytolytic activity toward tumor cells. To further enhance the cytolysis of HTLV-I-infected cells, we modified m8Δ and obtained m8Δ/RT1AlSCTax180L, which can express a single chain trimer (SCT) of rat major histocompatibility complex class I with a Tax-epitope. Combined treatment with m8Δ/RT1AlSCTax180L and 4O1/C8 increased the cytolysis of FPM1V.EFGFP/8R cells, a CTL-resistant subclone of FPM1, compared with that using 4O1/C8 and m8Δ presenting an unrelated peptide, suggesting that the activation of 4O1/C8 by m8Δ/RT1AlSCTax180L further enhanced the killing of the tumorigenic HTLV-I-infected cells. Our results indicate that combined therapy of oncolytic VVs with SCTs and HTLV-I-specific CTLs may be effective for eradication of HTLV-I-infected cells, which evade from CTL lysis and potentially develop ATL.
Collapse
|
1893
|
Gromov P, Moreira JMA, Gromova I. Proteomic analysis of tissue samples in translational breast cancer research. Expert Rev Proteomics 2014; 11:285-302. [DOI: 10.1586/14789450.2014.899469] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
1894
|
Kwak Y, Lee HE, Kim WH, Kim DW, Kang SB, Lee HS. The clinical implication of cancer-associated microvasculature and fibroblast in advanced colorectal cancer patients with synchronous or metachronous metastases. PLoS One 2014; 9:e91811. [PMID: 24642707 PMCID: PMC3958375 DOI: 10.1371/journal.pone.0091811] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 02/14/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND We aimed to evaluate the clinical significance of microvessel density (MVD), lymphatic vessel density (LVD), and cancer-associated fibroblasts (CAFs) in relation to tumor location in advanced colorectal cancer (CRC). METHODS Using immunohistochemistry, we examined 181 advanced CRC patients for CD31 and D2-40 to measure MVD and LVD, respectively, α-smooth muscle actin (SMA) and desmin to identify CAFs, and PTEN to examine genetic changes of CAFs. To evaluate the regional heterogeneity of these properties, we examined tissue from four sites (the center and periphery of the primary cancer, a distant metastasis, and a lymph node metastasis) in each patient. RESULTS MVD, LVD, and CAFs showed significant heterogeneity with respect to the tumor location. LVD was the greatest in the center of the primary cancers and the amount of CAFs was the lowest in distant metastases. In distant metastases, those from the lung had higher LVD and MVD, but fewer CAFs than those from the liver, peritoneum, or ovary. Patients with low MVD and LVD in the center of the primary cancer had worse outcomes and patients with few CAFs in distant metastases and in the primary tumor had a lower survival rate. PTEN expression in CAFs in distant metastases was lost in 11 of 181 CRC patients (6.1%), which was associated with a worse prognosis. CONCLUSIONS The microenvironment, including cancer-associated microvasculature and fibroblasts, is heterogeneous with respect to the tumor location in CRC patients. Therefore, heterogeneity of microenvironments should be taken into account when managing CRC patients.
Collapse
Affiliation(s)
- Yoonjin Kwak
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Eun Lee
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Duck-Woo Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
| | - Sung-Bum Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
| |
Collapse
|
1895
|
Botter SM, Neri D, Fuchs B. Recent advances in osteosarcoma. Curr Opin Pharmacol 2014; 16:15-23. [PMID: 24632219 DOI: 10.1016/j.coph.2014.02.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 01/09/2023]
Abstract
Although osteosarcoma (OS) is a rare malignancy, it is ranked among the leading causes of cancer-related death in the pediatric age group. The cancer's low prevalence and its large tumor heterogeneity make it difficult to obtain meaningful progress in patient survival. In this review we present an overview of current clinical trials which largely focus on stimulation of the immune system or rely on the inhibition of kinases such as Src and mTOR. The potential efficacy of tumor-targeted TNFalpha is discussed, as well as the importance of preclinical validation of new targets. To improve the success of future clinical trials, clinicians and basic researchers need to intensify their exchange. Finally, a case is made for individualized treatment of OS patients, based on interdisciplinary cooperation in dedicated Sarcoma Centers.
Collapse
Affiliation(s)
- Sander M Botter
- Sarcoma Center & Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | - Bruno Fuchs
- Sarcoma Center & Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland.
| |
Collapse
|
1896
|
Mittal K, Ebos J, Rini B. Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond. Semin Oncol 2014; 41:235-51. [PMID: 24787295 DOI: 10.1053/j.seminoncol.2014.02.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our understanding of the dynamic tumor microenvironment (TME) has improved exponentially over the last few decades. In addition to traditional cytotoxic agents, anti-cancer strategies now include numerous molecular-targeted drugs that modulate distinct elements of the TME. Angiogenesis is an underlying promoter of tumor growth, invasion, and metastases. From traditional and emerging angiogenic cytokines and their receptors to novel immune checkpoint inhibitors, regulation of the tumor microenvironment is potentially key in countering tumor progression. In this article, an overview of the architecture of the TME and the orchestration of angiogenesis within the TME is provided. Additionally, traditional and novel angiogenic targets of current interest within the TME are reviewed.
Collapse
Affiliation(s)
- Kriti Mittal
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195.
| | - John Ebos
- Roswell Park Cancer Institute, Buffalo, NY
| | - Brian Rini
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195
| |
Collapse
|
1897
|
Candia J, Banavar JR, Losert W. Understanding health and disease with multidimensional single-cell methods. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2014; 26:073102. [PMID: 24451406 PMCID: PMC4020281 DOI: 10.1088/0953-8984/26/7/073102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Current efforts in the biomedical sciences and related interdisciplinary fields are focused on gaining a molecular understanding of health and disease, which is a problem of daunting complexity that spans many orders of magnitude in characteristic length scales, from small molecules that regulate cell function to cell ensembles that form tissues and organs working together as an organism. In order to uncover the molecular nature of the emergent properties of a cell, it is essential to measure multiple-cell components simultaneously in the same cell. In turn, cell heterogeneity requires multiple-cells to be measured in order to understand health and disease in the organism. This review summarizes current efforts towards a data-driven framework that leverages single-cell technologies to build robust signatures of healthy and diseased phenotypes. While some approaches focus on multicolor flow cytometry data and other methods are designed to analyze high-content image-based screens, we emphasize the so-called Supercell/SVM paradigm (recently developed by the authors of this review and collaborators) as a unified framework that captures mesoscopic-scale emergence to build reliable phenotypes. Beyond their specific contributions to basic and translational biomedical research, these efforts illustrate, from a larger perspective, the powerful synergy that might be achieved from bringing together methods and ideas from statistical physics, data mining, and mathematics to solve the most pressing problems currently facing the life sciences.
Collapse
Affiliation(s)
- Julián Candia
- Department of Physics, University of Maryland, College Park, MD 20742, USA. School of Medicine, University of Maryland, Baltimore, MD 21201, USA. IFLYSIB and CONICET, University of La Plata, 1900 La Plata, Argentina
| | | | | |
Collapse
|
1898
|
Zhang J, O'Carroll SJ, Henare K, Ching LM, Ormonde S, Nicholson LFB, Danesh-Meyer HV, Green CR. Connexin hemichannel induced vascular leak suggests a new paradigm for cancer therapy. FEBS Lett 2014; 588:1365-71. [PMID: 24548560 DOI: 10.1016/j.febslet.2014.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 11/15/2022]
Abstract
It is 40 years since cancer growth was correlated with neovascularisation. Anti-angiogenic drugs remain at the forefront of cancer investigations but progress has been disappointing and unexpected toxicities are emerging. Gap junction channels are implicated in lesion spread following injury, with channel blockers shown to improve healing; in particular preventing vascular disruption and/or restoring vascular integrity. Here we briefly review connexin roles in vascular leak and endothelial cell death that occurs following acute wounds and during chronic disease, and how connexin channel regulation has been used to ameliorate vascular disruption. We then review chronic inflammatory disorders and trauma in the eye, concluding that vascular disruption under these conditions mimics that seen in tumours, and can be prevented with connexin hemichannel modulation. We apply this knowledge to tumour vessel biology, proposing that contrary to current opinion, these data suggest a need to protect, maintain and/or restore cancer vasculature. This may lead to reduced tumour hypoxia, promote the survival of normal cells, and enable improved therapeutic delivery or more effective radiation therapy.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Simon J O'Carroll
- Department of Anatomy and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Kimiora Henare
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Lai-Ming Ching
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Susan Ormonde
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Louise F B Nicholson
- Department of Anatomy and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Helen V Danesh-Meyer
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
1899
|
Li M, Xiao X, Zhang W, Liu L, Xi N, Wang Y. Nanoscale distribution of CD20 on B-cell lymphoma tumour cells and its potential role in the clinical efficacy of rituximab. J Microsc 2014; 254:19-30. [PMID: 24499016 DOI: 10.1111/jmi.12112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
Rituximab is an exciting monoclonal antibody drug approved for treating B-cell lymphomas and its target is the CD20 antigen which is expressed on the surface of B cells. In recent years, the variable efficacies of rituximab among different lymphoma patients have become an important clinical issue and urgently need to be solved for further development of antibodies with enhanced efficacies. In this work, atomic force microscopy (AFM) was used to investigate the nanoscale distribution of CD20 on the surface of tumour B cells from lymphoma patients to examine its potential role in the clinical therapeutic effects of rituximab. By performing ROR1 fluorescence labelling (ROR1 is a specific tumour cell surface marker) on the bone marrow cells prepared from B-cell lymphoma patients, the tumour B cells were recognized, and then AFM tips carrying rituximabs via polyethylene glycol crosslinkers were moved to the tumour cells to probe the specific CD20-rituximab interactions. By applying AFM single-molecule force spectroscopy (SMFS) at the local areas (500×500 nm²) on the surface of tumour B cells, the nanoscale distributions of CD20 on the surface of tumour B cells were mapped, visually showing that CD20 distributed heterogeneously on the cell surface. Bone marrow cell samples from three clinical B-cell lymphoma cases were collected to analyze the binding affinity and nanoscale distribution of CD20 on tumour cells. The experimental results showed that CD20 distribution on tumour cells were to some extent related to the clinical therapeutic outcomes while the CD20-rituximab binding forces did not have distinct effects to the clinical outcomes. These results can provide novel insights in understanding the rituximab's clinical efficacies from the nanoscale distribution of CD20 on the tumour cells at single-cell and single-molecule levels.
Collapse
Affiliation(s)
- M Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.,University of Chinese Academy of Sciences, Beijing, China
| | - X Xiao
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, China
| | - W Zhang
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, China
| | - L Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - N Xi
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.,Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Y Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| |
Collapse
|
1900
|
Cho YW, Kim SY, Kwon IC, Kim IS. Complex adaptive therapeutic strategy (CATS) for cancer. J Control Release 2014; 175:43-7. [DOI: 10.1016/j.jconrel.2013.12.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/19/2013] [Accepted: 12/16/2013] [Indexed: 12/16/2022]
|