1701
|
Asghar W, El Assal R, Shafiee H, Pitteri S, Paulmurugan R, Demirci U. Engineering cancer microenvironments for in vitro 3-D tumor models. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2015; 18:539-553. [PMID: 28458612 PMCID: PMC5407188 DOI: 10.1016/j.mattod.2015.05.002] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The natural microenvironment of tumors is composed of extracellular matrix (ECM), blood vasculature, and supporting stromal cells. The physical characteristics of ECM as well as the cellular components play a vital role in controlling cancer cell proliferation, apoptosis, metabolism, and differentiation. To mimic the tumor microenvironment outside the human body for drug testing, two-dimensional (2-D) and murine tumor models are routinely used. Although these conventional approaches are employed in preclinical studies, they still present challenges. For example, murine tumor models are expensive and difficult to adopt for routine drug screening. On the other hand, 2-D in vitro models are simple to perform, but they do not recapitulate natural tumor microenvironment, because they do not capture important three-dimensional (3-D) cell-cell, cell-matrix signaling pathways, and multi-cellular heterogeneous components of the tumor microenvironment such as stromal and immune cells. The three-dimensional (3-D) in vitro tumor models aim to closely mimic cancer microenvironments and have emerged as an alternative to routinely used methods for drug screening. Herein, we review recent advances in 3-D tumor model generation and highlight directions for future applications in drug testing.
Collapse
Affiliation(s)
- Waseem Asghar
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Computer Engineering & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Rami El Assal
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Hadi Shafiee
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Division of Biomedical Engineering, Division of Infectious Diseases, Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sharon Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Utkan Demirci
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Division of Biomedical Engineering, Division of Infectious Diseases, Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
1702
|
Seebacher NA, Lane DJR, Jansson PJ, Richardson DR. Glucose Modulation Induces Lysosome Formation and Increases Lysosomotropic Drug Sequestration via the P-Glycoprotein Drug Transporter. J Biol Chem 2015; 291:3796-820. [PMID: 26601947 DOI: 10.1074/jbc.m115.682450] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Indexed: 11/06/2022] Open
Abstract
Pgp is functional on the plasma membrane and lysosomal membrane. Lysosomal-Pgp can pump substrates into the organelle, thereby trapping certain chemotherapeutics (e.g. doxorubicin; DOX). This mechanism serves as a "safe house" to protect cells against cytotoxic drugs. Interestingly, in contrast to DOX, lysosomal sequestration of the novel anti-tumor agent and P-glycoprotein (Pgp) substrate, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), induces lysosomal membrane permeabilization. This mechanism of lysosomal-Pgp utilization enhances cytotoxicity to multidrug-resistant cells. Consequently, Dp44mT has greater anti-tumor activity in drug-resistant relative to non-Pgp-expressing tumors. Interestingly, stressors in the tumor microenvironment trigger endocytosis for cell signaling to assist cell survival. Hence, this investigation examined how glucose variation-induced stress regulated early endosome and lysosome formation via endocytosis of the plasma membrane. Furthermore, the impact of glucose variation-induced stress on resistance to DOX was compared with Dp44mT and its structurally related analogue, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC). These studies showed that glucose variation-induced stress-stimulated formation of early endosomes and lysosomes. In fact, through the process of fluid-phase endocytosis, Pgp was redistributed from the plasma membrane to the lysosomal membrane via early endosome formation. This lysosomal-Pgp actively transported the Pgp substrate, DOX, into the lysosome where it became trapped as a result of protonation at pH 5. Due to increased lysosomal DOX trapping, Pgp-expressing cells became more resistant to DOX. In contrast, cytotoxicity of Dp44mT and DpC was potentiated due to more lysosomes containing functional Pgp under glucose-induced stress. These thiosemicarbazones increased lysosomal membrane permeabilization and cell death. This mechanism has critical implications for drug-targeting in multidrug-resistant tumors where a stressful micro-environment exists.
Collapse
Affiliation(s)
- Nicole A Seebacher
- From the Molecular Pharmacology and Pathology Program, Department of Pathology, Blackburn Building (D06), University of Sydney, New South Wales, Australia 2006
| | - Darius J R Lane
- From the Molecular Pharmacology and Pathology Program, Department of Pathology, Blackburn Building (D06), University of Sydney, New South Wales, Australia 2006
| | - Patric J Jansson
- From the Molecular Pharmacology and Pathology Program, Department of Pathology, Blackburn Building (D06), University of Sydney, New South Wales, Australia 2006
| | - Des R Richardson
- From the Molecular Pharmacology and Pathology Program, Department of Pathology, Blackburn Building (D06), University of Sydney, New South Wales, Australia 2006
| |
Collapse
|
1703
|
Tinganelli W, Durante M, Hirayama R, Krämer M, Maier A, Kraft-Weyrather W, Furusawa Y, Friedrich T, Scifoni E. Kill-painting of hypoxic tumours in charged particle therapy. Sci Rep 2015; 5:17016. [PMID: 26596243 PMCID: PMC4657060 DOI: 10.1038/srep17016] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/23/2015] [Indexed: 02/07/2023] Open
Abstract
Solid tumours often present regions with severe oxygen deprivation (hypoxia), which
are resistant to both chemotherapy and radiotherapy. Increased radiosensitivity as a
function of the oxygen concentration is well described for X-rays. It has also been
demonstrated that radioresistance in anoxia is reduced using high-LET radiation
rather than conventional X-rays. However, the dependence of the oxygen enhancement
ratio (OER) on radiation quality in the regions of intermediate oxygen
concentrations, those normally found in tumours, had never been measured and
biophysical models were based on extrapolations. Here we present a complete survival
dataset of mammalian cells exposed to different ions in oxygen concentration ranging
from normoxia (21%) to anoxia (0%). The data were used to generate a model of the
dependence of the OER on oxygen concentration and particle energy. The model was
implemented in the ion beam treatment planning system to prescribe uniform cell
killing across volumes with heterogeneous radiosensitivity. The adaptive treatment
plans have been validated in two different accelerator facilities, using a
biological phantom where cells can be irradiated simultaneously at three different
oxygen concentrations. We thus realized a hypoxia-adapted treatment plan, which will
be used for painting by voxel of hypoxic tumours visualized by functional
imaging.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany.,Research Center for Charged Particle Therapy and International Open Laboratory, National Institute of Radiological Sciences, 263-8555 Chiba, Japan
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany.,Technical University Darmstadt, 64283 Darmstadt, Germany
| | - Ryoichi Hirayama
- Research Center for Charged Particle Therapy and International Open Laboratory, National Institute of Radiological Sciences, 263-8555 Chiba, Japan
| | - Michael Krämer
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
| | - Andreas Maier
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
| | - Wilma Kraft-Weyrather
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
| | - Yoshiya Furusawa
- Research Center for Charged Particle Therapy and International Open Laboratory, National Institute of Radiological Sciences, 263-8555 Chiba, Japan
| | - Thomas Friedrich
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
| | - Emanuele Scifoni
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
| |
Collapse
|
1704
|
Boonstra MC, Prakash J, Van De Velde CJH, Mesker WE, Kuppen PJK, Vahrmeijer AL, Sier CFM. Stromal Targets for Fluorescent-Guided Oncologic Surgery. Front Oncol 2015; 5:254. [PMID: 26636036 PMCID: PMC4653299 DOI: 10.3389/fonc.2015.00254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Pre-operative imaging techniques are essential for tumor detection and diagnosis, but offer limited help during surgery. Recently, the applicability of imaging during oncologic surgery has been recognized, using near-infrared fluorescent dyes conjugated to targeting antibodies, peptides, or other vehicles. Image-guided oncologic surgery (IGOS) assists the surgeFon to distinguish tumor from normal tissue during operation, and can aid in recognizing vital structures. IGOS relies on an optimized combination of a dedicated fluorescent camera system and specific probes for targeting. IGOS probes for clinical use are not widely available yet, but numerous pre-clinical studies have been published and clinical trials are being established or prepared. Most of the investigated probes are based on antibodies or peptides against proteins on the membranes of malignant cells, whereas others are directed against stromal cells. Targeting stroma cells for IGOS has several advantages. Besides the high stromal content in more aggressive tumor types, the stroma is often primarily located at the periphery/invasive front of the tumor, which makes stromal targets particularly suited for imaging purposes. Moreover, because stroma up-regulation is a physiological reaction, most proteins to be targeted on these cells are “universal” and not derived from a specific genetic variation, as is the case with many upregulated proteins on malignant cancer cells.
Collapse
Affiliation(s)
- Martin C Boonstra
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Jai Prakash
- Department of Biomaterial Science and Technology, Targeted Therapeutics, University of Twente , Enschede , Netherlands
| | | | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands ; Antibodies for Research Applications BV , Gouda , Netherlands
| |
Collapse
|
1705
|
de Velasco G, Krajewski KM, Albiges L, Awad MM, Bellmunt J, Hodi FS, Choueiri TK. Radiologic Heterogeneity in Responses to Anti-PD-1/PD-L1 Therapy in Metastatic Renal Cell Carcinoma. Cancer Immunol Res 2015; 4:12-7. [PMID: 26589768 DOI: 10.1158/2326-6066.cir-15-0197] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/21/2015] [Indexed: 11/16/2022]
Abstract
Radiologic assessment of tumor response remains a challenge in patients treated with immune checkpoint inhibitors. In metastatic melanoma, for example, a spectrum of imaging patterns in response to immunotherapies have been recognized and associated with clinical benefit. In metastatic renal cell carcinoma (mRCC), less than half of patients treated with immune checkpoint inhibitors achieve objective responses, but some of the responses have been durable. In this series, five different imaging patterns of response and progression are described in mRCC patients treated with anti-PD-1/PD-L1 agents: (i) early and complete response, (ii) pseudoprogression, (iii) disease stability before ultimate response, (iv) mixed response with new lesions, and (v) early progression/primary refractory disease. The implications of the different imaging patterns of patient responses on disease prognosis are discussed and highlight the need for individualized patient assessment when using these novel immune-targeted agents.
Collapse
Affiliation(s)
- Guillermo de Velasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laurence Albiges
- Department of Medical Oncology, Institut Gustave-Roussy, Villejuif, France
| | - Mark M Awad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Joaquim Bellmunt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
1706
|
Lan C, Heindl A, Huang X, Xi S, Banerjee S, Liu J, Yuan Y. Quantitative histology analysis of the ovarian tumour microenvironment. Sci Rep 2015; 5:16317. [PMID: 26573438 PMCID: PMC4647219 DOI: 10.1038/srep16317] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/12/2015] [Indexed: 11/09/2022] Open
Abstract
Concerted efforts in genomic studies examining RNA transcription and DNA methylation patterns have revealed profound insights in prognostic ovarian cancer subtypes. On the other hand, abundant histology slides have been generated to date, yet their uses remain very limited and largely qualitative. Our goal is to develop automated histology analysis as an alternative subtyping technology for ovarian cancer that is cost-efficient and does not rely on DNA quality. We developed an automated system for scoring primary tumour sections of 91 late-stage ovarian cancer to identify single cells. We demonstrated high accuracy of our system based on expert pathologists' scores (cancer = 97.1%, stromal = 89.1%) as well as compared to immunohistochemistry scoring (correlation = 0.87). The percentage of stromal cells in all cells is significantly associated with poor overall survival after controlling for clinical parameters including debulking status and age (multivariate analysis p = 0.0021, HR = 2.54, CI = 1.40-4.60) and progression-free survival (multivariate analysis p = 0.022, HR = 1.75, CI = 1.09-2.82). We demonstrate how automated image analysis enables objective quantification of microenvironmental composition of ovarian tumours. Our analysis reveals a strong effect of the tumour microenvironment on ovarian cancer progression and highlights the potential of therapeutic interventions that target the stromal compartment or cancer-stroma signalling in the stroma-high, late-stage ovarian cancer subset.
Collapse
Affiliation(s)
- Chunyan Lan
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Andreas Heindl
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Molecular Pathology, The Royal Marsden Hospital, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Xin Huang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Shaoyan Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Susana Banerjee
- Gynecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Jihong Liu
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Molecular Pathology, The Royal Marsden Hospital, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
1707
|
Nawaz S, Yuan Y. Computational pathology: Exploring the spatial dimension of tumor ecology. Cancer Lett 2015; 380:296-303. [PMID: 26592351 DOI: 10.1016/j.canlet.2015.11.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023]
Abstract
Tumors are evolving ecosystems where cancer subclones and the microenvironment interact. This is analogous to interaction dynamics between species in their natural habitats, which is a prime area of study in ecology. Spatial statistics are frequently used in ecological studies to infer complex relations including predator-prey, resource dependency and co-evolution. Recently, the emerging field of computational pathology has enabled high-throughput spatial analysis by using image processing to identify different cell types and their locations within histological tumor samples. We discuss how these data may be analyzed with spatial statistics used in ecology to reveal patterns and advance our understanding of ecological interactions occurring among cancer cells and their microenvironment.
Collapse
Affiliation(s)
- Sidra Nawaz
- Centre for Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK; Centre for Evolution and Cancer, Institute of Cancer Research, London SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Yinyin Yuan
- Centre for Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK; Centre for Evolution and Cancer, Institute of Cancer Research, London SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
1708
|
Nagaraj AB, Joseph P, DiFeo A. miRNAs as prognostic and therapeutic tools in epithelial ovarian cancer. Biomark Med 2015; 9:241-57. [PMID: 25731210 DOI: 10.2217/bmm.14.108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and is the fifth leading cause of cancer deaths in women. Developing adjuvant therapy to circumvent drug resistance represents an important aspect of current initiatives to improve survival in women with advanced EOC. A regulatory molecule that can act on multiple genes associated with a chemoresistant phenotype will be the ideal target for the development of therapeutics to overcome resistance and miRNAs constitute promising tools in this regard. In this review, we discuss the emerging role of miRNAs in regulating EOC phenotype with a focus on prognostic and therapeutic importance of miRNAs and the possibility of miRNA modulation as a tool to improve efficacy of chemotherapy in EOC.
Collapse
Affiliation(s)
- Anil Belur Nagaraj
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
1709
|
Sameen S, Barbuti R, Milazzo P, Cerone A, Del Re M, Danesi R. Mathematical modeling of drug resistance due to KRAS mutation in colorectal cancer. J Theor Biol 2015; 389:263-73. [PMID: 26551156 DOI: 10.1016/j.jtbi.2015.10.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 01/31/2023]
Abstract
The most challenging task in colorectal cancer research nowadays is to understand the development of acquired resistance to anti-EGFR drugs. The key reason for this problem is the KRAS mutations appearance after the treatment with monoclonal antibodies (moAb). Here we present a mathematical model for the analysis of KRAS mutations behavior in colorectal cancer with respect to moAb treatments. To evaluate the drug performance we have developed equations for two types of tumors cells, KRAS mutated and KRAS wild-type. Both tumor cell populations were treated with a combination of moAb and chemotherapy drugs. It was observed that even the minimal initial concentration of KRAS mutation before the treatment has the ability to make the tumor refractory to the treatment. Minor population of KRAS mutations has strong influence on large number of wild-type cells as well rendering them resistant to chemotherapy. Patient׳s immune responses are specifically taken into considerations and it is found that, in case of KRAS mutations, the immune strength does not affect medication efficacy. Finally, cetuximab (moAb) and irinotecan (chemotherapy) drugs are analyzed as first-line treatment of colorectal cancer with few KRAS mutated cells. Results show that this combined treatment could be only effective for patients with high immune strengths and it should not be recommended as first-line therapy for patients with moderate immune strengths or weak immune systems because of a potential risk of relapse, with KRAS mutant cells acquired resistance involved with them.
Collapse
Affiliation(s)
- Sheema Sameen
- Department of Informatics, University of Pisa, Italy.
| | | | - Paolo Milazzo
- Department of Informatics, University of Pisa, Italy
| | | | - Marzia Del Re
- Clinical Pharmacology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Romano Danesi
- Clinical Pharmacology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
1710
|
Owusu BY, Vaid M, Kaler P, Klampfer L. Prognostic and Predictive Significance of Stromal Fibroblasts and Macrophages in Colon Cancer. BIOMARKERS IN CANCER 2015; 7:29-37. [PMID: 26568685 PMCID: PMC4631158 DOI: 10.4137/bic.s25247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022]
Abstract
Colon cancer development and malignant progression are driven by genetic and epigenetic alterations in tumor cells and by factors from the tumor microenvironment. Cancer cells become reliant on the activity of specific oncogenes and on prosurvival and proliferative signals they receive from the abnormal environment they create and reside in. Accordingly, the response to anticancer therapy is determined by genetic and epigenetic changes that are intrinsic to tumor cells and by the factors present in the tumor microenvironment. Recent advances in the understanding of the involvement of the tumor microenvironment in tumor progression and therapeutic response are optimizing the application of prognostic and predictive factors in colon cancer. Moreover, new targets in the tumor microenvironment that are amenable to therapeutic intervention have been identified. Because stromal cells are with rare exceptions genetically stable, the tumor microenvironment has emerged as a preferred target for therapeutic drugs. In this review, we discuss the role of stromal fibroblasts and macrophages in colon cancer progression and in the response of colon cancer patients to therapy.
Collapse
Affiliation(s)
- Benjamin Y Owusu
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Mudit Vaid
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Pawan Kaler
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Lidija Klampfer
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| |
Collapse
|
1711
|
Park KM, Gerecht S. Polymeric hydrogels as artificial extracellular microenvironments for cancer research. Eur Polym J 2015. [DOI: 10.1016/j.eurpolymj.2015.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
1712
|
Kwon MC, Berns A. Tumor heterogeneity-induced signaling regulates SCLC metastasis. Cell Cycle 2015; 14:3347-8. [PMID: 26506985 DOI: 10.1080/15384101.2015.1093445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Min-chul Kwon
- a Division of Molecular Genetics; The Netherlands Cancer Institute ; Amsterdam , The Netherlands
| | - Anton Berns
- a Division of Molecular Genetics; The Netherlands Cancer Institute ; Amsterdam , The Netherlands.,b Skolkovo Institute of Science and Technology; Skolkovo Innovation Center ; Moscow , Russia
| |
Collapse
|
1713
|
Mannavola F, Tucci M, Felici C, Stucci S, Silvestris F. miRNAs in melanoma: a defined role in tumor progression and metastasis. Expert Rev Clin Immunol 2015; 12:79-89. [PMID: 26505837 DOI: 10.1586/1744666x.2016.1100965] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The crosstalk of melanoma cells with components of the microenvironment promotes malignant cell proliferation and spread to distant tissues. Although the major pathogenetic events have already been elucidated, the mechanisms that drive the metastatic behavior of tumor cells are still undefined. MicroRNAs (miRNAs) are small non-coding RNAs that control post-transcriptional gene expression through interconnected kinases upstream of functional genes involved in tumor progression. Here, we review the biological relevance of melanoma-related miRNAs and focus on their potential role in propagating signals that may cause tumor microenvironment rearrangements, as well as disablement of the immune system and melanoma cell proliferation.
Collapse
Affiliation(s)
- Francesco Mannavola
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Marco Tucci
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Claudia Felici
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Stefania Stucci
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Franco Silvestris
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| |
Collapse
|
1714
|
Song W, Tang Z, Zhang D, Li M, Gu J, Chen X. A cooperative polymeric platform for tumor-targeted drug delivery. Chem Sci 2015; 7:728-736. [PMID: 28791115 PMCID: PMC5530016 DOI: 10.1039/c5sc01698c] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 10/22/2015] [Indexed: 12/30/2022] Open
Abstract
A tumor-targeted drug delivery system with small-molecule vascular disrupting agents inducing coagulation environment inside tumor and coagulation-targeted nanoparticles accumulating there.
In the pursuit of effective treatments for cancer, an emerging strategy is “active targeting”, where nanoparticles are decorated with targeting ligands able to recognize and bind specific receptors overexpressed by tumor cells or tumor vasculature so that a greater fraction of the administered drugs are selectively trafficked to tumor sites. However, the implementation of this strategy has faced a major obstacle. The interpatient, inter- and intra-tumoral heterogeneity in receptor expression can pose challenges for the design of clinical trials and result in the paucity of targetable receptors within a tumor, which limits the effectiveness of “active targeting” strategy in cancer treatment. Here we report a cooperative drug delivery platform that overcomes the heterogeneity barrier unique to solid tumors. The cooperative platform comprises a coagulation-inducing agent and coagulation-targeted polymeric nanoparticles. As a typical small-molecule vascular disrupting agent (VDA), DMXAA can create a unique artificial coagulation environment with additional binding sites in a solid tumor by locally activating a coagulation cascade. Coagulation-targeted cisplatin-loaded nanoparticles, which are surface-decorated with a substrate of activated blood coagulation factor XIII, can selectively accumulate in the solid tumor by homing to the VDA-induced artificial coagulation environment through transglutamination. In vivo studies show that the cooperative tumor-selective platform recruits up to 7.5-fold increases in therapeutic cargos to the tumors and decreases tumor burden with low systemic toxicity as compared with non-cooperative controls. These indicate that the use of coagulation-targeted nanoparticles, in conjunction with free small-molecule VDAs, may be a valuable strategy for improving standard chemotherapy.
Collapse
Affiliation(s)
- Wantong Song
- Key Laboratory of Polymer Ecomaterials , Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , P. R. China . ;
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials , Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , P. R. China . ;
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials , Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , P. R. China . ;
| | - Mingqiang Li
- Key Laboratory of Polymer Ecomaterials , Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , P. R. China . ;
| | - Jingkai Gu
- Research Center for Drug Metabolism , College of Life Science , Jilin University , Changchun, 130012 , P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials , Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , P. R. China . ;
| |
Collapse
|
1715
|
Micro Regional Heterogeneity of 64Cu-ATSM and 18F-FDG Uptake in Canine Soft Tissue Sarcomas: Relation to Cell Proliferation, Hypoxia and Glycolysis. PLoS One 2015; 10:e0141379. [PMID: 26501874 PMCID: PMC4621038 DOI: 10.1371/journal.pone.0141379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Tumour microenvironment heterogeneity is believed to play a key role in cancer progression and therapy resistance. However, little is known about micro regional distribution of hypoxia, glycolysis and proliferation in spontaneous solid tumours. The overall aim was simultaneous investigation of micro regional heterogeneity of 64Cu-ATSM (hypoxia) and 18F-FDG (glycolysis) uptake and correlation to endogenous markers of hypoxia, glycolysis, proliferation and angiogenesis to better therapeutically target aggressive tumour regions and prognosticate outcome. METHODS Exploiting the different half-lives of 64Cu-ATSM (13 h) and 18F-FDG (2 h) enabled simultaneous investigation of micro regional distribution of hypoxia and glycolysis in 145 tumour pieces from four spontaneous canine soft tissue sarcomas. Pairwise measurements of radioactivity and gene expression of endogenous markers of hypoxia (HIF-1α, CAIX), glycolysis (HK2, GLUT1 and GLUT3), proliferation (Ki-67) and angiogenesis (VEGFA and TF) were performed. Dual tracer autoradiography was compared with Ki-67 immunohistochemistry. RESULTS Micro regional heterogeneity in hypoxia and glycolysis within and between tumour sections of each tumour piece was observed. The spatial distribution of 64Cu-ATSM and 18F-FDG was rather similar within each tumour section as reflected in moderate positive significant correlations between the two tracers (ρ = 0.3920-0.7807; p = 0.0180 -<0.0001) based on pixel-to-pixel comparisons of autoradiographies and gamma counting of tumour pieces. 64Cu-ATSM and 18F-FDG correlated positively with gene expression of GLUT1 and GLUT3, but negatively with HIF-1α and CAIX. Significant positive correlations were seen between Ki-67 gene expression and 64Cu-ATSM (ρ = 0.5578, p = 0.0004) and 18F-FDG (ρ = 0.4629-0.7001, p = 0.0001-0.0151). Ki-67 gene expression more consistently correlated with 18F-FDG than with 64Cu-ATSM. CONCLUSIONS Micro regional heterogeneity of hypoxia and glycolysis was documented in spontaneous canine soft tissue sarcomas. 64Cu-ATSM and 18F-FDG uptakes and distributions showed significant moderate correlations at the micro regional level indicating overlapping, yet different information from the tracers.18F-FDG better reflected cell proliferation as measured by Ki-67 gene expression than 64Cu-ATSM.
Collapse
|
1716
|
Boonzaier NR, Piccirillo SGM, Watts C, Price SJ. Assessing and monitoring intratumor heterogeneity in glioblastoma: how far has multimodal imaging come? CNS Oncol 2015; 4:399-410. [PMID: 26497327 DOI: 10.2217/cns.15.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma demonstrates imaging features of intratumor heterogeneity that result from underlying heterogeneous biological properties. This stems from variations in cellular behavior that result from genetic mutations that either drive, or are driven by, heterogeneous microenvironment conditions. Among all imaging methods available, only T1-weighted contrast-enhancing and T2-weighted fluid-attenuated inversion recovery are used in standard clinical glioblastoma assessment and monitoring. Advanced imaging modalities are still considered emerging techniques as appropriate end points and robust methodologies are missing from clinical trials. Discovering how these images specifically relate to the underlying tumor biology may aid in improving quality of clinical trials and understanding the factors involved in regional responses to treatment, including variable drug uptake and effect of radiotherapy. Upon validation and standardization of emerging MR techniques, providing information based on the underlying tumor biology, these images may allow for clinical decision-making that is tailored to an individual's response to treatment.
Collapse
Affiliation(s)
- Natalie R Boonzaier
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Sara G M Piccirillo
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Colin Watts
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Stephen J Price
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
1717
|
Chang J, Vacher J, Yao B, Fan X, Zhang B, Harris RC, Zhang MZ. Prostaglandin E receptor 4 (EP4) promotes colonic tumorigenesis. Oncotarget 2015; 6:33500-11. [PMID: 26378024 PMCID: PMC4741781 DOI: 10.18632/oncotarget.5589] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/23/2015] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) continues to be a major cause of morbidity and mortality. Although the factors underlying CRC development and progression are multifactorial, there is an important role for tumor-host interactions, especially interactions with myeloid cells. There is also increasing evidence that cyclooxygenase-derived prostaglandins are important mediators of CRC development and growth. Although prevention trials with either nonselective NSAIDs or COX-2 selective agents have shown promise, the gastrointestinal or cardiovascular side effects of these agents have limited their implementation. The predominant prostaglandin involved in CRC pathogenesis is PGE2. Since myeloid cells express high levels of the PGE2 receptor subtype, EP4, we selectively ablated EP4 in myeloid cells and studied adenoma formation in a mouse model of intestinal adenomatous polyposis, ApcMin/+ mice. ApcMin/+mice with selective myeloid cell deletion of EP4 had marked inhibition of both adenoma number and size, with associated decreases in mTOR and ERK activation. Either genetic or pharmacologic inhibition of EP4 receptors led to an anti-tumorigenic M1 phenotype of macrophages/dendritic cells. Therefore, PGE2-mediated EP4 signaling in myeloid cells promotes tumorigenesis, suggesting EP4 as a potentially attractive target for CRC chemoprevention or treatment.
Collapse
Affiliation(s)
- Jian Chang
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hepatobiliary Surgery Department, Wuhan No.1 Hospital, Wuhan, China
| | - Jean Vacher
- Départment of Médecine, Clinical Research Institute of Montreal, Université de Montréal, Montreal, Quebec, Canada
| | - Bing Yao
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Xiaofeng Fan
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Raymond C. Harris
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Jiangsu Center for The Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, China
| |
Collapse
|
1718
|
Enomoto M, Vaughen J, Igaki T. Non-autonomous overgrowth by oncogenic niche cells: Cellular cooperation and competition in tumorigenesis. Cancer Sci 2015; 106:1651-8. [PMID: 26362609 PMCID: PMC4714670 DOI: 10.1111/cas.12816] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/23/2022] Open
Abstract
Tumor progression is classically viewed as the Darwinian evolution of subclones that sequentially acquire genetic mutations and autonomously overproliferate. However, growing evidence suggests that tumor microenvironment and subclone heterogeneity contribute to non‐autonomous tumor progression. Recent Drosophila studies revealed a common mechanism by which clones of genetically altered cells trigger non‐autonomous overgrowth. Such “oncogenic niche cells” (ONCs) do not overgrow but instead stimulate neighbor overgrowth and metastasis. Establishment of ONCs depends on competition and cooperation between heterogeneous cell populations. This review characterizes diverse ONCs identified in Drosophila and describes the genetic basis of non‐autonomous tumor progression. Similar mechanisms may contribute to mammalian cancer progression and recurrence.
Collapse
Affiliation(s)
- Masato Enomoto
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - John Vaughen
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
1719
|
An in vitro assessment of liposomal topotecan simulating metronomic chemotherapy in combination with radiation in tumor-endothelial spheroids. Sci Rep 2015; 5:15236. [PMID: 26468877 PMCID: PMC4606561 DOI: 10.1038/srep15236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 09/02/2015] [Indexed: 12/31/2022] Open
Abstract
Low dose metronomic chemotherapy (LDMC) refers to prolonged administration of low dose chemotherapy designed to minimize toxicity and target the tumor endothelium, causing tumor growth inhibition. Topotecan (TPT) when administered at its maximum tolerated dose (MTD) is often associated with systemic hematological toxicities. Liposomal encapsulation of TPT enhances efficacy by shielding it from systemic clearance, allowing greater uptake and extended tissue exposure in tumors. Extended release of TPT from liposomal formulations also has the potential to mimic metronomic therapies with fewer treatments. Here we investigate potential toxicities of equivalent doses of free and actively loaded liposomal TPT (LTPT) and compare them to a fractionated low dose regimen of free TPT in tumor-endothelial spheroids (TES) with/without radiation exposure for a prolonged period of 10 days. Using confocal microscopy, TPT fluorescence was monitored to determine the accumulation of drug within TES. These studies showed TES, being more reflective of the in vivo tumor microenvironment, were more sensitive to LTPT in comparison to free TPT with radiation. More importantly, the response of TES to low-dose metronomic TPT with radiation was comparable to similar treatment with LTPT. This TES study suggests nanoparticle formulations designed for extended release of drug can simulate LDMC in vivo.
Collapse
|
1720
|
Yamaguchi H, Sakai R. Direct Interaction between Carcinoma Cells and Cancer Associated Fibroblasts for the Regulation of Cancer Invasion. Cancers (Basel) 2015; 7:2054-62. [PMID: 26473929 PMCID: PMC4695876 DOI: 10.3390/cancers7040876] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 09/30/2015] [Accepted: 10/12/2015] [Indexed: 12/14/2022] Open
Abstract
The tumor stroma acts as an essential microenvironment of the cancer cells, which includes many different types of non-cancerous cells and the extracellular matrix (ECM). Stromal fibroblasts (SFs) are the major cellular constituents of the tumor stroma and are often called cancer-associated fibroblasts (CAFs). They are often characterized by α-smooth muscle actin (αSMA) expression, which is indicative of the myofibroblast phenotype and strong contractility. These characteristics contribute to the remodeling and stiffening of the stromal ECM, thereby offering an appropriate field for cancer cell invasion. Importance of the tumor stroma in cancer progression has recently been highlighted. Moreover, several reports suggest that stromal fibroblasts interact with adjacent cancer cells through soluble factors, exosomes, or direct cell-cell adhesion to promote cancer cell invasion. In this review, current models of the regulation of cancer cell invasion by surrounding fibroblasts are summarized, including our recent work on the interaction between stromal fibroblasts and scirrhous gastric carcinoma (SGC) cells by using a three-dimensional (3D) culture system. Further mechanistic insights into the roles of the interaction between cancer cells and stromal fibroblasts in cancer invasion will be required to identify novel molecular targets for preventing cancer cell invasion.
Collapse
Affiliation(s)
- Hideki Yamaguchi
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Ryuichi Sakai
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
- Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan.
| |
Collapse
|
1721
|
Pathological responses after angiogenesis or EGFR inhibitors in metastatic colorectal cancer depend on the chemotherapy backbone. Br J Cancer 2015; 113:1298-304. [PMID: 26461062 PMCID: PMC4815793 DOI: 10.1038/bjc.2015.321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 07/25/2015] [Accepted: 08/12/2015] [Indexed: 12/22/2022] Open
Abstract
Background: Optimal preoperative treatment before colorectal cancer metastases (CRCM) resection remains unclear. This study evaluated pathological responses (pR) in CRCM resected after chemotherapy alone or combined with angiogenesis or epidermal growth factor receptor (EGFR) inhibitors. Methods: Pathological response was retrospectively evaluated on 264 resected metastases from 99 patients. The proportion of responding metastases after different preoperative treatments was reported and compared. Patient's progression-free survival (PFS) and overall survival (OS) were compared based on pR. Results: The combination of anti-angiogenics with oxaliplatin-based chemotherapy resulted in more pR than when they were combined with irinotecan-based chemotherapy (80% vs 50% P<0.001). Inversely, the combination of EGFR inhibitors with oxaliplatin-based chemotherapy seemed to induce fewer pR than when they were combined with irinotecan-based treatment (53% vs 72% P=0.049). Overall survival at 5 years was improved for patients with a pR in all resected metastases compared with those who did not achieve a pR (68.5% vs 32.6% P=0.023) and this response was the only factor predicting OS in a multivariate analysis. Conclusion: The chemotherapy partner combined with angiogenesis or EGFR inhibitors influenced pR in resected CRCM. In our exploratory analysis anti-angiogenic/oxaliplatin-based regimens and anti-EGFR/irinotecan-based regimens were associated with the highest pR. Prospective randomised trials should be performed to validate these observations.
Collapse
|
1722
|
Drug-repositioning opportunities for cancer therapy: novel molecular targets for known compounds. Drug Discov Today 2015; 21:190-199. [PMID: 26456577 DOI: 10.1016/j.drudis.2015.09.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/21/2015] [Accepted: 09/30/2015] [Indexed: 01/10/2023]
Abstract
Drug repositioning is gaining increasing attention in drug discovery because it represents a smart way to exploit new molecular targets of a known drug or target promiscuity among diverse diseases, for medical uses different from the one originally considered. In this review, we focus on known non-oncological drugs with new therapeutic applications in oncology, explaining the rationale behind this approach and providing practical evidence. Moving from incompleteness of the knowledge of drug-target interactions, particularly for older molecules, we highlight opportunities for repurposing compounds as cancer therapeutics, underling the biologically and clinically relevant affinities for new targets. Ideal candidates for repositioning can contribute to the therapeutically unmet need for more-efficient anticancer agents, including drugs that selectively target cancer stem cells.
Collapse
|
1723
|
Reichel D, Rychahou P, Bae Y. Polymer nanoassemblies with solvato- and halo-fluorochromism for drug release monitoring and metastasis imaging. Ther Deliv 2015; 6:1221-37. [PMID: 26446432 PMCID: PMC4977001 DOI: 10.4155/tde.15.59] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Theranostics, an emerging technique that combines therapeutic and diagnostic modalities for various diseases, holds promise to detect cancer in early stages, eradicate metastatic tumors and ultimately reduce cancer mortality. METHODS & RESULTS This study reports unique polymer nanoassemblies that increase fluorescence intensity upon addition of hydrophobic drugs and either increase or decrease fluorescence intensity in acidic environments, depending on nanoparticle core environment properties. Extensive spectroscopic analyses were performed to determine optimal excitation and emission wavelengths, which enabled real time measurement of drugs releasing from the nanoassemblies and ex vivo imaging of acidic liver metastatic tumors from mice. CONCLUSION Polymer nanoassemblies with solvato- and halo-fluorochromic properties are promising platforms to develop novel theranostic tools for the detection and treatment of metastatic tumors.
Collapse
Affiliation(s)
- Derek Reichel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, KY 40536–0596, USA
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, 800 Rose Street, CC140, Lexington, KY 40536, USA
- Department of Surgery, College of Medicine, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, KY 40536–0596, USA
| |
Collapse
|
1724
|
Custead MR, An R, Turek JJ, Moore GE, Nolte DD, Childress MO. Predictive value of ex vivo biodynamic imaging in determining response to chemotherapy in dogs with spontaneous non-Hodgkin's lymphomas: a preliminary study. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2015; 1. [PMID: 27280042 DOI: 10.1088/2057-1739/1/1/015003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Biodynamic imaging (BDI) is a novel phenotypic cancer profiling technology which optically characterizes changes in subcellular motion within living tumor tissue samples in response to ex vivo treatment with cancer chemotherapy drugs. The purpose of this preliminary study was to assess the ability of ex vivo BDI to predict in vivo clinical response to chemotherapy in ten dogs with naturally-occurring non-Hodgkin's lymphomas. Pre-treatment tumor biopsy samples were obtained from all dogs and treated ex vivo with doxorubicin (10 μM). BDI measured six dynamic biomarkers of subcellular motion from all biopsy samples at baseline and at regular intervals for 9 h following drug application. All dogs subsequently received doxorubicin to treat their lymphomas. Best overall response to and progression-free survival time following chemotherapy were recorded for all dogs. Receiver operating characteristic (ROC) curves were used to determine accuracy and identify possible cut-off values for the BDI-measured biomarkers which could accurately predict those dogs' cancers that would and would not respond to doxorubicin chemotherapy. One biomarker (designated 'MEM') showed 100% discriminative capability for predicting clinical response to doxorubicin (area under the ROC curve = 1.00, 95% CI 0.692-1.000), while other biomarkers also showed promising predictive capability. These preliminary findings suggest that ex vivo BDI can accurately predict treatment outcome following doxorubicin chemotherapy in a spontaneous animal cancer model, and is worthy of further investigation as a technology for personalized cancer medicine.
Collapse
Affiliation(s)
- M R Custead
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| | - R An
- Department of Physics and Astronomy, Purdue University Department of Physics and Astronomy, Purdue University, 525 Northwestern Avenue, West Lafayette, IN 47907-2036, USA
| | - J J Turek
- Department of Basic Medical Sciences, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA.,Purdue University Center for Cancer Research, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| | - G E Moore
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| | - D D Nolte
- Department of Physics and Astronomy, Purdue University Department of Physics and Astronomy, Purdue University, 525 Northwestern Avenue, West Lafayette, IN 47907-2036, USA.,Purdue University Center for Cancer Research, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| | - M O Childress
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA.,Purdue University Center for Cancer Research, Purdue University College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| |
Collapse
|
1725
|
Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:111. [PMID: 26445347 PMCID: PMC4595070 DOI: 10.1186/s13046-015-0221-y] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022]
Abstract
Tumor tissue is composed of cancer cells and surrounding stromal cells with diverse genetic/epigenetic backgrounds, a situation known as intra-tumoral heterogeneity. Cancer cells are surrounded by a totally different microenvironment than that of normal cells; consequently, tumor cells must exhibit rapidly adaptive responses to hypoxia and hypo-nutrient conditions. This phenomenon of changes of tumor cellular bioenergetics, called “metabolic reprogramming”, has been recognized as one of 10 hallmarks of cancer. Metabolic reprogramming is required for both malignant transformation and tumor development, including invasion and metastasis. Although the Warburg effect has been widely accepted as a common feature of metabolic reprogramming, accumulating evidence has revealed that tumor cells depend on mitochondrial metabolism as well as aerobic glycolysis. Remarkably, cancer-associated fibroblasts in tumor stroma tend to activate both glycolysis and autophagy in contrast to neighboring cancer cells, which leads to a reverse Warburg effect. Heterogeneity of monocarboxylate transporter expression reflects cellular metabolic heterogeneity with respect to the production and uptake of lactate. In tumor tissue, metabolic heterogeneity induces metabolic symbiosis, which is responsible for adaptation to drastic changes in the nutrient microenvironment resulting from chemotherapy. In addition, metabolic heterogeneity is responsible for the failure to induce the same therapeutic effect against cancer cells as a whole. In particular, cancer stem cells exhibit several biological features responsible for resistance to conventional anti-tumor therapies. Consequently, cancer stem cells tend to form minimal residual disease after chemotherapy and exhibit metastatic potential with additional metabolic reprogramming. This type of altered metabolic reprogramming leads to adaptive/acquired resistance to anti-tumor therapy. Collectively, complex and dynamic metabolic reprogramming should be regarded as a reflection of the “robustness” of tumor cells against unfavorable conditions. This review focuses on the concept of metabolic reprogramming in heterogeneous tumor tissue, and further emphasizes the importance of developing novel therapeutic strategies based on drug repositioning.
Collapse
Affiliation(s)
- Go J Yoshida
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan. .,Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
1726
|
Islam F, Gopalan V, Wahab R, Smith RA, Lam AKY. Cancer stem cells in oesophageal squamous cell carcinoma: Identification, prognostic and treatment perspectives. Crit Rev Oncol Hematol 2015; 96:9-19. [PMID: 25913844 DOI: 10.1016/j.critrevonc.2015.04.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 03/03/2015] [Accepted: 04/07/2015] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a vital subpopulation of cells to target for the treatment of cancers. In oesophageal squamous cell carcinoma (ESCC), there are several markers such as CD44, ALDH, Pygo2, MAML1, Twist1, Musashi1, Side population (SP), CD271 and CD90 that have been proposed to identify the cancer stem cells in individual cancer masses. It has also been demonstrated that stem cell markers like ALDH1, HIWI, Oct3/4, ABCG2, SOX2, SALL4, BMI-1, NANOG, CD133 and podoplanin are associated with patient's prognosis, pathological stages, cancer recurrence and therapy resistance. Finding new cancer stem cell targets or designing drugs to manipulate the known molecular targets in CSCs could be useful for improvements in clinical outcomes of the disease. To conclude, data suggest that CSCs in oesophageal squamous cell carcinoma are related to resistance to therapy and poor prognosis of patients with ESCC. Therefore, innovative insights into CSC biology and CSC-targeted therapies will help to achieve more effective management of patients with oesophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Riajul Wahab
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Robert A Smith
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Alfred K-Y Lam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
1727
|
Muhanna N, Mepham A, Mohamadi RM, Chan H, Khan T, Akens M, Besant JD, Irish J, Kelley SO. Nanoparticle-based sorting of circulating tumor cells by epithelial antigen expression during disease progression in an animal model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1613-20. [DOI: 10.1016/j.nano.2015.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/17/2015] [Accepted: 04/25/2015] [Indexed: 12/29/2022]
|
1728
|
Intra-tumor heterogeneity of cancer cells and its implications for cancer treatment. Acta Pharmacol Sin 2015; 36:1219-27. [PMID: 26388155 PMCID: PMC4648179 DOI: 10.1038/aps.2015.92] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/06/2015] [Indexed: 02/06/2023]
Abstract
Recent studies have revealed extensive genetic and non-genetic variation across different geographical regions of a tumor or throughout different stages of tumor progression, which is referred to as intra-tumor heterogeneity. Several causes contribute to this phenomenon, including genomic instability, epigenetic alteration, plastic gene expression, signal transduction, and microenvironmental differences. These variables may affect key signaling pathways that regulate cancer cell growth, drive phenotypic diversity, and pose challenges to cancer treatment. Understanding the mechanisms underlying this heterogeneity will support the development of effective therapeutic strategies.
Collapse
|
1729
|
Ugel S, Facciponte JG, De Sanctis F, Facciabene A. Targeting tumor vasculature: expanding the potential of DNA cancer vaccines. Cancer Immunol Immunother 2015; 64:1339-48. [PMID: 26267042 PMCID: PMC11028665 DOI: 10.1007/s00262-015-1747-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/28/2015] [Indexed: 01/16/2023]
Abstract
Targeting the tumor vasculature with anti-angiogenesis modalities is a bona fide validated approach that has complemented cancer treatment paradigms. Tumor vasculature antigens (TVA) can be immunologically targeted and offers multiple theoretical advantages that may enhance existing strategies against cancer. We focused on tumor endothelial marker 1 (TEM1/CD248) as a model TVA since it is broadly expressed on many different cancers. Our DNA-based vaccine approach demonstrated that CD248 can be effectively targeted immunologically; anti-tumor responses were generated in several mouse models; and CD8(+)/CD4(+) T cell responses were elicited against peptides derived from CD248 protein. Our work supports our contention that CD248 is a novel immunotherapeutic target for cancer treatment and highlights the efficient, safe and translatable use of DNA-based immunotherapy. We next briefly highlight ongoing investigations targeting CD248 with antibodies as a diagnostic imaging agent and as a therapeutic antibody in an early clinical trial. The optimal approach for generating effective DNA-based cancer vaccines for several tumor types may be a combinatorial approach that enhances immunogenicity such as combination with chemotherapy. Additional combination approaches are discussed and include those that alleviate the immunosuppressive tumor microenvironment induced by myeloid-derived suppressor cells and T regulatory cells. Targeting the tumor vasculature by CD248-based immunological modalities expands the armamentarium against cancer.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/therapeutic use
- Combined Modality Therapy
- Disease Models, Animal
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Humans
- Immunotherapy/methods
- Neoplasms/immunology
- Neoplasms/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- T-Lymphocytes/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Stefano Ugel
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - John G. Facciponte
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
| | - Francesco De Sanctis
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Andrea Facciabene
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
| |
Collapse
|
1730
|
Yang Q, Parker CL, McCallen JD, Lai SK. Addressing challenges of heterogeneous tumor treatment through bispecific protein-mediated pretargeted drug delivery. J Control Release 2015; 220:715-26. [PMID: 26407672 DOI: 10.1016/j.jconrel.2015.09.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 01/02/2023]
Abstract
Tumors are frequently characterized by genomically and phenotypically distinct cancer cell subpopulations within the same tumor or between tumor lesions, a phenomenon termed tumor heterogeneity. These diverse cancer cell populations pose a major challenge to targeted delivery of diagnostic and/or therapeutic agents, as the conventional approach of conjugating individual ligands to nanoparticles is often unable to facilitate intracellular delivery to the full spectrum of cancer cells present in a given tumor lesion or patient. As a result, many cancers are only partially suppressed, leading to eventual tumor regrowth and/or the development of drug-resistant tumors. Pretargeting (multistep targeting) approaches involving the administration of 1) a cocktail of bispecific proteins that can collectively bind to the entirety of a mixed tumor population followed by 2) nanoparticles containing therapeutic and/or diagnostic agents that can bind to the bispecific proteins accumulated on the surface of target cells offer the potential to overcome many of the challenges associated with drug delivery to heterogeneous tumors. Despite its considerable success in improving the efficacy of radioimmunotherapy, the pretargeting strategy remains underexplored for a majority of nanoparticle therapeutic applications, especially for targeted delivery to heterogeneous tumors. In this review, we will present concepts in tumor heterogeneity, the shortcomings of conventional targeted systems, lessons learned from pretargeted radioimmunotherapy, and important considerations for harnessing the pretargeting strategy to improve nanoparticle delivery to heterogeneous tumors.
Collapse
Affiliation(s)
- Qi Yang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Christina L Parker
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Justin D McCallen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Samuel K Lai
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, United States.
| |
Collapse
|
1731
|
Apostoli AJ, Ailles L. Clonal evolution and tumor-initiating cells: New dimensions in cancer patient treatment. Crit Rev Clin Lab Sci 2015; 53:40-51. [PMID: 26397062 DOI: 10.3109/10408363.2015.1083944] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human cancer is not a uniform disease but a plethora of disparate tumor types and subtypes. The differences that exist between individual tumors (intertumoral heterogeneity) present a significant roadblock to the eradication of cancer. It has also become increasingly clear that variations across individual tumors (intratumoral heterogeneity) have important implications to cancer progression and treatment efficacy. Therefore, in order to improve patient care and develop novel chemotherapeutics, the evolving tumor landscape needs to be further explored. Next-generation sequencing (NGS) technologies are revolutionizing the cancer research arena by providing state-of-the-art, high-speed methods of genome sequencing at single-nucleotide resolution, thus enabling an unprecedented detection of tumor-specific genetic abnormalities. These anomalies can be quantified to reveal specific frequencies of DNA alterations that correspond to distinct clonal populations within a given tumor. As such, NGS approaches have also been utilized to explore the heterogeneous landscape of patient tumors as well as to match metastatic and/or recurrent growths and patient-derived engrafts. By sequencing in this manner--through time so to speak--cancer researchers can track shifting clonal populations, make important inferences about tumor evolution and potentially identify tumor subclones that could be viably targeted. This exciting new territory has important implications for the competing clonal evolution and cancer stem cell models of tumor heterogeneity, and also offers a new dimension for cancer treatment and profound hope for patients in the coming years.
Collapse
Affiliation(s)
- Anthony J Apostoli
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and
| | - Laurie Ailles
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and.,b Department of Medical Biophysics , University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|
1732
|
Weiswald LB, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. Neoplasia 2015; 17:1-15. [PMID: 25622895 PMCID: PMC4309685 DOI: 10.1016/j.neo.2014.12.004] [Citation(s) in RCA: 815] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 11/29/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional (3D) in vitro models have been used in cancer research as an intermediate model between in vitro cancer cell line cultures and in vivo tumor. Spherical cancer models represent major 3D in vitro models that have been described over the past 4 decades. These models have gained popularity in cancer stem cell research using tumorospheres. Thus, it is crucial to define and clarify the different spherical cancer models thus far described. Here, we focus on in vitro multicellular spheres used in cancer research. All these spherelike structures are characterized by their well-rounded shape, the presence of cancer cells, and their capacity to be maintained as free-floating cultures. We propose a rational classification of the four most commonly used spherical cancer models in cancer research based on culture methods for obtaining them and on subsequent differences in sphere biology: the multicellular tumor spheroid model, first described in the early 70s and obtained by culture of cancer cell lines under nonadherent conditions; tumorospheres, a model of cancer stem cell expansion established in a serum-free medium supplemented with growth factors; tissue-derived tumor spheres and organotypic multicellular spheroids, obtained by tumor tissue mechanical dissociation and cutting. In addition, we describe their applications to and interest in cancer research; in particular, we describe their contribution to chemoresistance, radioresistance, tumorigenicity, and invasion and migration studies. Although these models share a common 3D conformation, each displays its own intrinsic properties. Therefore, the most relevant spherical cancer model must be carefully selected, as a function of the study aim and cancer type.
Collapse
Affiliation(s)
- Louis-Bastien Weiswald
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Laboratoire d'Oncobiologie, Hôpital René Huguenin, Institut Curie, St Cloud, France; Université Paris Descartes, Faculté de Pharmacie de Paris, Sorbonne Paris Cité, Paris, France.
| | - Dominique Bellet
- Laboratoire d'Oncobiologie, Hôpital René Huguenin, Institut Curie, St Cloud, France; Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, UMR 8151 CNRS-U1022 Inserm, Sorbonne Paris Cité, Paris, France
| | - Virginie Dangles-Marie
- Université Paris Descartes, Faculté de Pharmacie de Paris, Sorbonne Paris Cité, Paris, France; Département de Recherche Translationnelle, Research Center, Institut Curie, Paris, France.
| |
Collapse
|
1733
|
Yang F, Yu Y. [Tumor microenvironment--the critical element of tumor metastasis]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:48-54. [PMID: 25603873 PMCID: PMC5999737 DOI: 10.3779/j.issn.1009-3419.2015.01.08] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
肿瘤转移是癌症治疗失败和患者死亡的主要原因,其分子机制复杂,涉及多步骤、多阶段、多基因的变化。作为肿瘤细胞赖以生存的场所,肿瘤微环境在肿瘤转移过程中起到至关重要的作用。因此,研究肿瘤微环境与肿瘤转移的动态关系,阐明微环境中不同因子在转移过程中的分子机制是抑制肿瘤转移的关键。
Collapse
Affiliation(s)
- Fang Yang
- Department of Medical Oncology, Tumor Hospital, Harbin Medical University, Harbin 150081, China
| | - Yan Yu
- Department of Medical Oncology, Tumor Hospital, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
1734
|
Li M, Liu L, Xi N, Wang Y, Xiao X, Zhang W. Effects of temperature and cellular interactions on the mechanics and morphology of human cancer cells investigated by atomic force microscopy. SCIENCE CHINA-LIFE SCIENCES 2015; 58:889-901. [PMID: 26354505 DOI: 10.1007/s11427-015-4914-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 05/06/2015] [Indexed: 12/19/2022]
Abstract
Cell mechanics plays an important role in cellular physiological activities. Recent studies have shown that cellular mechanical properties are novel biomarkers for indicating the cell states. In this article, temperature-controllable atomic force microscopy (AFM) was applied to quantitatively investigate the effects of temperature and cellular interactions on the mechanics and morphology of human cancer cells. First, AFM indenting experiments were performed on six types of human cells to investigate the changes of cellular Young's modulus at different temperatures and the results showed that the mechanical responses to the changes of temperature were variable for different types of cancer cells. Second, AFM imaging experiments were performed to observe the morphological changes in living cells at different temperatures and the results showed the significant changes of cell morphology caused by the alterations of temperature. Finally, by co-culturing human cancer cells with human immune cells, the mechanical and morphological changes in cancer cells were investigated. The results showed that the co-culture of cancer cells and immune cells could cause the distinct mechanical changes in cancer cells, but no significant morphological differences were observed. The experimental results improved our understanding of the effects of temperature and cellular interactions on the mechanics and morphology of cancer cells.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - LianQing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
| | - Ning Xi
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China. .,Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| | - YueChao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
| | - XiuBin Xiao
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, 100071, China
| | - WeiJing Zhang
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, 100071, China
| |
Collapse
|
1735
|
Jorth P, Staudinger BJ, Wu X, Hisert KB, Hayden H, Garudathri J, Harding CL, Radey MC, Rezayat A, Bautista G, Berrington WR, Goddard AF, Zheng C, Angermeyer A, Brittnacher MJ, Kitzman J, Shendure J, Fligner CL, Mittler J, Aitken ML, Manoil C, Bruce JE, Yahr TL, Singh PK. Regional Isolation Drives Bacterial Diversification within Cystic Fibrosis Lungs. Cell Host Microbe 2015; 18:307-19. [PMID: 26299432 PMCID: PMC4589543 DOI: 10.1016/j.chom.2015.07.006] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 06/25/2015] [Accepted: 07/16/2015] [Indexed: 12/22/2022]
Abstract
Bacterial lineages that chronically infect cystic fibrosis (CF) patients genetically diversify during infection. However, the mechanisms driving diversification are unknown. By dissecting ten CF lung pairs and studying ∼12,000 regional isolates, we were able to investigate whether clonally related Pseudomonas aeruginosa inhabiting different lung regions evolve independently and differ functionally. Phylogenetic analysis of genome sequences showed that regional isolation of P. aeruginosa drives divergent evolution. We investigated the consequences of regional evolution by studying isolates from mildly and severely diseased lung regions and found evolved differences in bacterial nutritional requirements, host defense and antibiotic resistance, and virulence due to hyperactivity of the type 3 secretion system. These findings suggest that bacterial intermixing is limited in CF lungs and that regional selective pressures may markedly differ. The findings also may explain how specialized bacterial variants arise during infection and raise the possibility that pathogen diversification occurs in other chronic infections characterized by spatially heterogeneous conditions.
Collapse
Affiliation(s)
- Peter Jorth
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Benjamin J Staudinger
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Xia Wu
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Katherine B Hisert
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Hillary Hayden
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jayanthi Garudathri
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Christopher L Harding
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Matthew C Radey
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Amir Rezayat
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Gilbert Bautista
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William R Berrington
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Amanda F Goddard
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Chunxiang Zheng
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Angus Angermeyer
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Mitchell J Brittnacher
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jacob Kitzman
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Corinne L Fligner
- Departments of Pathology and Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - John Mittler
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Moira L Aitken
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Colin Manoil
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - James E Bruce
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Timothy L Yahr
- Department of Microbiology, University of Iowa School of Medicine, Iowa City, IA 52240, USA
| | - Pradeep K Singh
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
1736
|
Physical Intimacy of Breast Cancer Cells with Mesenchymal Stem Cells Elicits Trastuzumab Resistance through Src Activation. Sci Rep 2015; 5:13744. [PMID: 26345302 PMCID: PMC4561910 DOI: 10.1038/srep13744] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 07/17/2015] [Indexed: 12/27/2022] Open
Abstract
The development of resistance to trastuzumab is a major obstacle for lasting effective treatment of patients with ErbB2-overexpressing tumors. Here, we demonstrate that the physical contact of breast cancer cells with mesenchymal stem cells (MSCs) is a potential modulator of trastuzumab response by activation of nonreceptor tyrosine kinase c-Src and down regulation of phosphatase and tensin homolog (PTEN). Using an in vitro patterned breast cancer/MSC co-culture model, we find that the presence of MSCs results in Src activation that is missing in cancer cells monoculture, transwell co-culture, and cells treated with MSCs conditioned media. Interestingly, the co-culture model also results in PTEN loss and activation of PI3K/AKT pathway that has been demonstrated as fundamental proliferative and survival pathways in clinical settings. To our knowledge, this is the first report that showed PTEN loss without the use of chemical inhibitors, matrix stiffness, or silencing RNAs. In addition, breast cancer cells in co-culture with MSCs conferred trastuzumab resistance in vitro as observed in the lack of inhibition of proliferative and migrative properties of the cancer cells. Our findings show that MSCs are potent mediators of resistance to trastuzumab and might reveal targets to enhance trastuzumab efficacy in patients.
Collapse
|
1737
|
Valeta-Magara A, Hatami R, Axelrod D, Roses DF, Guth A, Formenti SC, Schneider RJ. Pro-oncogenic cytokines and growth factors are differentially expressed in the post-surgical wound fluid from malignant compared to benign breast lesions. SPRINGERPLUS 2015; 4:483. [PMID: 26361584 PMCID: PMC4560730 DOI: 10.1186/s40064-015-1260-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 08/18/2015] [Indexed: 12/16/2022]
Abstract
Purpose The accumulation of wound fluid known as seroma in the chest cavity following breast surgery is a common occurrence that can persist for many weeks. While the pro-inflammatory composition of seroma is well established, there has been remarkably little research to determine whether seroma contains pro-oncogenic factors, and whether this is influenced by previous malignant disease. Methods We developed a clinical trial in which we obtained post-surgical seroma fluids from women with benign or malignant disease 1 or 2 weeks following lumpectomy or mastectomy. We conducted an analysis of more than 80 different cytokines, chemokines and growth factors. Results We found that surgical cavity seroma from breast cancer patients has a higher expression of key tumor-promoting cytokines and lower expression of important tumor-inhibiting factors when compared to benign lesions from non-cancer patients. Patients with high body mass index also had higher levels of leptin regardless of malignancy. Conclusions We conclude that the breast post-surgical tumor cavity contains factors that are pro-inflammatory regardless of malignant or benign disease, but in malignant disease there is significant enrichment of additional pro-oncogenic chemokines, cytokines and growth factors, and reduction in tumor-inhibiting factors. These results are consistent with tumor conditioning of surrounding normal stromal tissue and creation of a pro-oncogenic environment that persists long after surgical removal of the tumor. Electronic supplementary material The online version of this article (doi:10.1186/s40064-015-1260-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Raheleh Hatami
- Department of Microbiology, NYU School of Medicine, New York, NY 10016 USA
| | - Deborah Axelrod
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Daniel F Roses
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Amber Guth
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Silvia C Formenti
- Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016 USA ; Weill Cornell Medical College, New York, NY USA
| | - Robert J Schneider
- Department of Microbiology, NYU School of Medicine, New York, NY 10016 USA ; Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA ; Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016 USA
| |
Collapse
|
1738
|
Kolch W, Halasz M, Granovskaya M, Kholodenko BN. The dynamic control of signal transduction networks in cancer cells. Nat Rev Cancer 2015; 15:515-27. [PMID: 26289315 DOI: 10.1038/nrc3983] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is often considered a genetic disease. However, much of the enormous plasticity of cancer cells to evolve different phenotypes, to adapt to challenging microenvironments and to withstand therapeutic assaults is encoded by the structure and spatiotemporal dynamics of signal transduction networks. In this Review, we discuss recent concepts concerning how the rich signalling dynamics afforded by these networks are regulated and how they impinge on cancer cell proliferation, survival, invasiveness and drug resistance. Understanding this dynamic circuitry by mathematical modelling could pave the way to new therapeutic approaches and personalized treatments.
Collapse
Affiliation(s)
- Walter Kolch
- Systems Biology Ireland, University College Dublin
- Conway Institute of Biomolecular &Biomedical Research, University College Dublin
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Marina Granovskaya
- Roche Moscow Limited, Business Center Neglinnaya Plaza, Building 2, Trubnaya Square, 107031 Moscow, Russia
| | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin
- Conway Institute of Biomolecular &Biomedical Research, University College Dublin
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
1739
|
Shahar E, Gorodetsky R, Aizenshtein E, Lalush L, Pitcovski J. Modulating the innate immune activity in murine tumor microenvironment by a combination of inducer molecules attached to microparticles. Cancer Immunol Immunother 2015; 64:1137-49. [PMID: 26031575 PMCID: PMC11028936 DOI: 10.1007/s00262-015-1719-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
Targeted cancer immunotherapy is challenging due to the cellular diversity and imposed immune tolerance in the tumor microenvironment (TME). A promising route to overcome those drawbacks may be by activating innate immune cells (IIC) in the TME, toward tumor destruction. Studies have shown the ability to "re-educate" pro-tumor-activated IIC toward antitumor responses. The current research aims to stimulate such activation using a combination of innate activators loaded onto microparticles (MP). Four inducers of Toll-like receptors 4 and 7, complement C5a receptor (C5aR) and gamma Fc receptor and their combinations were loaded on MP, and their influence on immune cell activation evaluated. MP stimulation of immune cell activation was tested in vitro and in vivo using a subcutaneous B16-F10 melanoma model induced in C57BL6 mice. Exposure to the TLR4 ligand lipopolysaccharide (LPS) bound to MP-induced acute inflammatory cytokine and chemokine activity in vitro and in vivo, with the elevation of CD45(+) leukocytes in particular GR-1(+) neutrophils and F4/80 macrophages in the TME. Nevertheless, LPS alone on MP was insufficient to significantly delay tumor progression. LPS combined with the C5aR ligand C5a-pep on the same MP resulted in a similar inflammation activation pattern. However, interleukin-10 levels were lower, and tumor growth was significantly delayed. Mixtures of these two ligands on separate MP did not yield the same cytokine activation pattern, demonstrating the importance of the cells' dual activation. The results suggest that combining inducers of distinct innate immune activation pathways holds promise for successful redirection of TME-residing IIC toward anti-tumoral activation.
Collapse
Affiliation(s)
- Ehud Shahar
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Lab of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah – Hebrew University Medical Center, Jerusalem, Israel
| | - Raphael Gorodetsky
- Lab of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah – Hebrew University Medical Center, Jerusalem, Israel
| | - Elina Aizenshtein
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
| | - Lior Lalush
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Tel Hai Academic College, Upper Galilee, Israel
| | - Jacob Pitcovski
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Tel Hai Academic College, Upper Galilee, Israel
| |
Collapse
|
1740
|
Maris P, Blomme A, Palacios AP, Costanza B, Bellahcène A, Bianchi E, Gofflot S, Drion P, Trombino GE, Di Valentin E, Cusumano PG, Maweja S, Jerusalem G, Delvenne P, Lifrange E, Castronovo V, Turtoi A. Asporin Is a Fibroblast-Derived TGF-β1 Inhibitor and a Tumor Suppressor Associated with Good Prognosis in Breast Cancer. PLoS Med 2015; 12:e1001871. [PMID: 26327350 PMCID: PMC4556693 DOI: 10.1371/journal.pmed.1001871] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/21/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Breast cancer is a leading malignancy affecting the female population worldwide. Most morbidity is caused by metastases that remain incurable to date. TGF-β1 has been identified as a key driving force behind metastatic breast cancer, with promising therapeutic implications. METHODS AND FINDINGS Employing immunohistochemistry (IHC) analysis, we report, to our knowledge for the first time, that asporin is overexpressed in the stroma of most human breast cancers and is not expressed in normal breast tissue. In vitro, asporin is secreted by breast fibroblasts upon exposure to conditioned medium from some but not all human breast cancer cells. While hormone receptor (HR) positive cells cause strong asporin expression, triple-negative breast cancer (TNBC) cells suppress it. Further, our findings show that soluble IL-1β, secreted by TNBC cells, is responsible for inhibiting asporin in normal and cancer-associated fibroblasts. Using recombinant protein, as well as a synthetic peptide fragment, we demonstrate the ability of asporin to inhibit TGF-β1-mediated SMAD2 phosphorylation, epithelial to mesenchymal transition, and stemness in breast cancer cells. In two in vivo murine models of TNBC, we observed that tumors expressing asporin exhibit significantly reduced growth (2-fold; p = 0.01) and metastatic properties (3-fold; p = 0.045). A retrospective IHC study performed on human breast carcinoma (n = 180) demonstrates that asporin expression is lowest in TNBC and HER2+ tumors, while HR+ tumors have significantly higher asporin expression (4-fold; p = 0.001). Assessment of asporin expression and patient outcome (n = 60; 10-y follow-up) shows that low protein levels in the primary breast lesion significantly delineate patients with bad outcome regardless of the tumor HR status (area under the curve = 0.87; 95% CI 0.78-0.96; p = 0.0001). Survival analysis, based on gene expression (n = 375; 25-y follow-up), confirmed that low asporin levels are associated with a reduced likelihood of survival (hazard ratio = 0.58; 95% CI 0.37-0.91; p = 0.017). Although these data highlight the potential of asporin to serve as a prognostic marker, confirmation of the clinical value would require a prospective study on a much larger patient cohort. CONCLUSIONS Our data show that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. High asporin expression is significantly associated with less aggressive tumors, stratifying patients according to the clinical outcome. Future pre-clinical studies should consider options for increasing asporin expression in TNBC as a promising strategy for targeted therapy.
Collapse
Affiliation(s)
- Pamela Maris
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Ana Perez Palacios
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Brunella Costanza
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | | | - Pierre Drion
- Animal Facility, GIGA–Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Giovanna Elvi Trombino
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | | | - Pino G. Cusumano
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Sylvie Maweja
- Department of Abdominal Surgery, University of Liège, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Eric Lifrange
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| |
Collapse
|
1741
|
Liu JY, Li F, Wang LP, Chen XF, Wang D, Cao L, Ping Y, Zhao S, Li B, Thorne SH, Zhang B, Kalinski P, Zhang Y. CTL- vs Treg lymphocyte-attracting chemokines, CCL4 and CCL20, are strong reciprocal predictive markers for survival of patients with oesophageal squamous cell carcinoma. Br J Cancer 2015; 113:747-755. [PMID: 26284335 PMCID: PMC4559838 DOI: 10.1038/bjc.2015.290] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/14/2015] [Accepted: 07/20/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Tumoural infiltration of T lymphocytes is determined by local patterns of specific chemokine expression. In this report, we examined the roles of CCL4 and CCL20 in the accumulation of CD8(+) cytotoxic T lymphocytes (CTLs) and regulatory T (Treg) lymphocytes in oesophageal squamous cell carcinoma (ESCC), and determined the correlations between chemokine expressions and ESCC patients' survival. METHODS Reverse transcriptase-PCR and immunohistochemistry (IHC) staining were performed to examine the expressions of interested genes. Flow cytometry was adopted to check the expressions of CCL4- and CCL6-specific receptors, CCR5 and CCR6, on CTLs and Treg cells. In addition, transwell assay was carried on. RESULTS The CCL4 expression was significantly correlated with the expression of CTL markers (CD8 and Granzyme B), whereas CCL20 was positively correlated with Treg markers (FoxP3 and IL-10). Consistently, CCR5 was found to be mainly expressed on CD8(+) T lymphocytes, while CCR6 showed prevalence on Treg lymphocytes and the frequencies of CCR5(+)CD8(+) CTLs and CCR6(+) Treg cells were higher in TIL compared with PBMC. Respectively, CCL4 and CCL20 recruited CD8(+) and regulatory T cells in vitro. Importantly, high levels of CCL4 in the lesions of ESCC patients predicted prolonged survival. Furthermore, CCL4(high)/CCL20(low) group demonstrated better overall survival, whereas CCL4(low)/CCL20(low) and CCL4(low)/CCL20(high) groups showed the worst overall survival. CONCLUSIONS Our data showed that CCL4 and CCL20 recruit functionally different T lymphocyte subsets into oesophageal carcinoma, indicating CCL4 and CCL20 are potential predictors of ESCC patients' survival.
Collapse
Affiliation(s)
- J Y Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - F Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - L P Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - X F Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - D Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - L Cao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Y Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - S Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - B Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - S H Thorne
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - B Zhang
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - P Kalinski
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Y Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan 450052, China
| |
Collapse
|
1742
|
Gandellini P, Andriani F, Merlino G, D'Aiuto F, Roz L, Callari M. Complexity in the tumour microenvironment: Cancer associated fibroblast gene expression patterns identify both common and unique features of tumour-stroma crosstalk across cancer types. Semin Cancer Biol 2015; 35:96-106. [PMID: 26320408 DOI: 10.1016/j.semcancer.2015.08.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/17/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Cancer is a complex disease, driven by the accumulation of several somatic aberrations but fostered by a two-way interaction between tumour cells and the surrounding microenvironment. Cancer associated fibroblasts (CAFs) represent one of the major players in tumour-stroma crosstalk. Recent in vitro and in vivo studies, often conducted by employing high throughput approaches, have started unravelling the key pathways involved in their functional effects. This review focus on open challenges in the study of CAF properties and function, highlighting at the same time the existence of common mechanisms as well as peculiarities in different cancer types (breast, prostate and lung cancer). Although still limited by current experimental models, which are unable to deal with the full level of complexity of the tumour microenvironment, a better understanding of these mechanisms may enable the identification of new biomarkers and therapeutic targets, to improve current strategies for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Paolo Gandellini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Andriani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca D'Aiuto
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Callari
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
1743
|
Reppas AI, Alfonso JCL, Hatzikirou H. In silico tumor control induced via alternating immunostimulating and immunosuppressive phases. Virulence 2015; 7:174-86. [PMID: 26305801 DOI: 10.1080/21505594.2015.1076614] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Despite recent advances in the field of Oncoimmunology, the success potential of immunomodulatory therapies against cancer remains to be elucidated. One of the reasons is the lack of understanding on the complex interplay between tumor growth dynamics and the associated immune system responses. Toward this goal, we consider a mathematical model of vascularized tumor growth and the corresponding effector cell recruitment dynamics. Bifurcation analysis allows for the exploration of model's dynamic behavior and the determination of these parameter regimes that result in immune-mediated tumor control. In this work, we focus on a particular tumor evasion regime that involves tumor and effector cell concentration oscillations of slowly increasing and decreasing amplitude, respectively. Considering a temporal multiscale analysis, we derive an analytically tractable mapping of model solutions onto a weakly negatively damped harmonic oscillator. Based on our analysis, we propose a theory-driven intervention strategy involving immunostimulating and immunosuppressive phases to induce long-term tumor control.
Collapse
Affiliation(s)
- A I Reppas
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| | - J C L Alfonso
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| | - H Hatzikirou
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| |
Collapse
|
1744
|
Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation. Nat Cell Biol 2015; 17:1193-204. [PMID: 26302407 PMCID: PMC4699446 DOI: 10.1038/ncb3228] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 07/20/2015] [Indexed: 12/13/2022]
Abstract
Stromal fibroblast senescence has been linked to aging-associated cancer risk. However, density and proliferation of cancer-associated fibroblasts (CAF) are frequently increased. Loss or down-modulation of the Notch effector CSL/RBP-Jκ in dermal fibroblasts is sufficient for CAF activation and ensuing keratinocyte-derived tumors. We report that CSL silencing induces senescence of primary fibroblasts from dermis, oral mucosa, breast and lung. CSL functions in these cells as direct repressor of multiple senescence- and CAF-effector genes. It also physically interacts with p53, repressing its activity. CSL is down-modulated in stromal fibroblasts of premalignant skin actinic keratosis lesions and squamous cell carcinomas (SCC), while p53 expression and function is down-modulated only in the latter, with paracrine FGF signaling as likely culprit. Concomitant loss of CSL and p53 overcomes fibroblast senescence, enhances expression of CAF effectors and promotes stromal and cancer cell expansion. The findings support a CAF activation/stromal co-evolution model under convergent CSL/p53 control.
Collapse
|
1745
|
Chiang YT, Gout PW, Collins CC, Wang Y. Prostate cancer metastasis-driving genes: hurdles and potential approaches in their identification. Asian J Androl 2015; 16:545-8. [PMID: 24589457 PMCID: PMC4104078 DOI: 10.4103/1008-682x.122875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Metastatic prostate cancer is currently incurable. Metastasis is thought to result from changes in the expression of specific metastasis-driving genes in nonmetastatic prostate cancer tissue, leading to a cascade of activated downstream genes that set the metastatic process in motion. Such genes could potentially serve as effective therapeutic targets for improved management of the disease. They could be identified by comparative analysis of gene expression profiles of patient-derived metastatic and nonmetastatic prostate cancer tissues to pinpoint genes showing altered expression, followed by determining whether silencing of such genes can lead to inhibition of metastatic properties. Various hurdles encountered in this approach are discussed, including (i) the need for clinically relevant, nonmetastatic and metastatic prostate cancer tissues such as xenografts of patients’ prostate cancers developed via subrenal capsule grafting technology and (ii) limitations in the currently available methodology for identification of master regulatory genes.
Collapse
Affiliation(s)
| | | | | | - Yuzhuo Wang
- The Vancouver Prostate Centre, Vancouver General Hospital and Department of Urologic Sciences, the University of British Columbia; Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| |
Collapse
|
1746
|
Khan S, Jaggi M, Chauhan SC. Revisiting stroma in pancreatic cancer. Oncoscience 2015; 2:819-20. [PMID: 26682261 PMCID: PMC4671936 DOI: 10.18632/oncoscience.198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023] Open
Affiliation(s)
- Sheema Khan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
1747
|
Foster JG, Wong SCK, Sharp TV. The hypoxic tumor microenvironment: driving the tumorigenesis of non-small-cell lung cancer. Future Oncol 2015; 10:2659-74. [PMID: 25531051 DOI: 10.2217/fon.14.201] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Since the application of molecular biology in cancer biology, lung cancer research has classically focused on molecular drivers of disease. One such pathway, the hypoxic response pathway, is activated by reduced local oxygen concentrations at the tumor site. Hypoxia-driven gene and protein changes enhance epithelial-to-mesenchymal transition, remodel the extracellular matrix, drive drug resistance, support cancer stem cells and aid evasion from immune cells. However, it is not the tumor cells alone which drive this response to hypoxia, but rather their interaction with a complex milieu of supporting cells. This review will focus on recent advances in our understanding of how these cells contribute to the tumor response to hypoxia in non-small-cell lung cancer.
Collapse
Affiliation(s)
- John G Foster
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | | | | |
Collapse
|
1748
|
Hui L, Chen Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett 2015; 368:7-13. [PMID: 26276713 DOI: 10.1016/j.canlet.2015.07.039] [Citation(s) in RCA: 555] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/28/2015] [Accepted: 07/16/2015] [Indexed: 12/24/2022]
Abstract
Tumor cells constantly interact with the surrounding microenvironment. Increasing evidence indicates that targeting the tumor microenvironment could complement traditional treatment and improve therapeutic outcomes for these malignancies. In this paper, we review new insights into the tumor microenvironment, and summarize selected examples of the cross-talk between tumor cells and their microenvironment, which have enhanced our understanding of pathophysiology of the microenvironment. We believe that this rapidly moving field promises many more to come, and they will guide the rational design of combinational therapies for success in cancer eradication.
Collapse
Affiliation(s)
- Lanlan Hui
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ye Chen
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China.
| |
Collapse
|
1749
|
Sun Y. Tumor microenvironment and cancer therapy resistance. Cancer Lett 2015; 380:205-15. [PMID: 26272180 DOI: 10.1016/j.canlet.2015.07.044] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/08/2015] [Accepted: 07/16/2015] [Indexed: 12/19/2022]
Abstract
Innate resistance to various therapeutic interventions is a hallmark of cancer. In recent years, however, acquired resistance has emerged as a daunting challenge to anticancer treatments including chemotherapy, radiation and targeted therapy, which abolishes the efficacy of otherwise successful regimens. Cancer cells gain resistance through a variety of mechanisms in both primary and metastatic sites, involving cell intrinsic and extrinsic factors, but the latter often remains overlooked. Mounting evidence suggests critical roles played by the tumor microenvironment (TME) in multiple aspects of cancer progression particularly therapeutic resistance. The TME decreases drug penetration, confers proliferative and antiapoptotic advantages to surviving cells, facilitates resistance without causing genetic mutations and epigenetic changes, collectively modifying disease modality and distorting clinical indices. Recent studies have set the baseline for future investigation on the intricate relationship between cancer resistance and the TME in pathological backgrounds. This review provides an updated outline of research advances in TME biology and highlights the prospect of targeting the TME as an essential strategy to overcome cancer resistance and improve therapeutic outcomes through precise intervention. In the long run, continued inputs into translational medicine remain highly desired to achieve durable responses in the current era of personalized clinical oncology.
Collapse
Affiliation(s)
- Yu Sun
- Key Lab of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai 200031, China; Collaborative Innovation Center of Systems Biomedicine, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China; VA Seattle Medical Center, Seattle, WA 98108, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
1750
|
He XP, Hu XL, Jin HY, Gan J, Zhu H, Li J, Long YT, Tian H. Quick Serological Detection of a Cancer Biomarker with an Agglutinated Supramolecular Glycoprobe. Anal Chem 2015; 87:9078-83. [PMID: 26235567 DOI: 10.1021/acs.analchem.5b02384] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Xiao-Peng He
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Xi-Le Hu
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Hong-Ying Jin
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Jiemin Gan
- Huadong Hospital Affiliated to Fudan University, 221 West Yan’an Road, Shanghai 200040, PR China
| | - Huili Zhu
- Huadong Hospital Affiliated to Fudan University, 221 West Yan’an Road, Shanghai 200040, PR China
| | - Jia Li
- National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Road, Shanghai 201203, PR China
| | - Yi-Tao Long
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - He Tian
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| |
Collapse
|