201
|
Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, Reyes-Soto G, Ortiz-Sánchez E, Herrera-Montalvo LA. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med 2020; 18:1. [PMID: 31900168 PMCID: PMC6941297 DOI: 10.1186/s12967-019-02189-8] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
The present review aimed to discuss contemporary scientific literature involving differences between the tumor microenvironment (TME) in melanoma, lung cancer, and breast cancer in their primary site and TME in brain metastases (BM). TME plays a fundamental role in the behavior of cancer. In the process of carcinogenesis, cells such as fibroblasts, macrophages, endothelial cells, natural killer cells, and other cells can perpetuate and progress carcinogenesis via the secretion of molecules. Oxygen concentration, growth factors, and receptors in TME initiate angiogenesis and are examples of the importance of microenvironmental conditions in the performance of neoplastic cells. The most frequent malignant brain tumors are metastatic in origin and primarily originate from lung cancer, breast cancer, and melanoma. Metastatic cancer cells have to adhere to and penetrate the blood-brain barrier (BBB). After traversing BBB, these cells have to survive by producing various cytokines, chemokines, and mediators to modify their new TME. The microenvironment of these metastases is currently being studied owing to the discovery of new therapeutic targets. In these three types of tumors, treatment is more effective in the primary tumor than in BM due to several factors, including BBB. Understanding the differences in the characteristics of the microenvironment surrounding the primary tumor and their respective metastasis might help improve strategies to comprehend cancer.
Collapse
Affiliation(s)
- Bernardo Cacho-Díaz
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico.
| | - Donovan R García-Botello
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Talia Wegman-Ostrosky
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Gervith Reyes-Soto
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Elizabeth Ortiz-Sánchez
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Luis Alonso Herrera-Montalvo
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico.
| |
Collapse
|
202
|
Izzi V, Koivunen J, Rappu P, Heino J, Pihlajaniemi T. Integration of Matrisome Omics: Towards System Biology of the Tumor Matrisome. EXTRACELLULAR MATRIX OMICS 2020. [DOI: 10.1007/978-3-030-58330-9_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
203
|
The Many Microenvironments of Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:199-213. [PMID: 34185294 DOI: 10.1007/978-3-030-59038-3_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most common and deadly subtype of ovarian cancer as it is commonly diagnosed after substantial metastasis has already occurred. The past two decades have been an active era in HGSOC research, with new information on the origin and genomic signature of the tumor cell. Additionally, studies have begun to characterize changes in the HGSOC microenvironment and examine the impact of these changes on tumor progression and response to therapies. While this knowledge may provide valuable insight into better prognosis and treatments for HGSOCs, its collection, synthesis, and application are complicated by the number of unique microenvironments in the disease-the initiating site (fallopian tube), first metastasis (ovary), distal metastases (peritoneum), and recurrent/platinum-resistant setting. Here, we review the state of our understanding of these diverse sites and highlight remaining questions.
Collapse
|
204
|
Ren Q, Yang K, Zou R, Wan Z, Shen Z, Wu G, Zhou Z, Ni Q, Fan W, Hu J, Liu Y. Biodegradable hollow manganese/cobalt oxide nanoparticles for tumor theranostics. NANOSCALE 2019; 11:23021-23026. [PMID: 31774090 DOI: 10.1039/c9nr07725a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This article describes the fabrication of hollow manganese/cobalt oxide nanoparticles (MCO NPs) with a tunable size through a redox reaction and the Kirkendall effect for cancer imaging and drug delivery. MCO-70 NPs (with an average size of 70 nm) can act as glutathione (GSH)-responsive contrast agents for dual T1/T2-weighted magnetic resonance imaging (MRI). The degradation of MCO NPs by GSH led to the enhancement of their T1 and T2 signals by 2.24- and 3.43-fold compared with those of MCO NPs before degradation, respectively. Antitumor agents such as doxorubicin (Dox) could be encapsulated inside the cavity of the hollow MCO NPs (MCO-70-Dox) and be released in the presence of GSH. The MCO-70-Dox NPs showed good tumor growth inhibition effects in vitro and in vivo, and can be promising drug delivery vehicles and MRI contrast agents for tumor diagnosis and reporting drug release.
Collapse
Affiliation(s)
- Qilong Ren
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Kuikun Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Rujia Zou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Zhiping Wan
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Zheyu Shen
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Guangyu Wu
- College of Biology and the Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Qianqian Ni
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Junqing Hu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Yijing Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
205
|
Sanchez A, Furberg H, Kuo F, Vuong L, Ged Y, Patil S, Ostrovnaya I, Petruzella S, Reising A, Patel P, Mano R, Coleman J, Russo P, Liu CH, Dannenberg AJ, Chan TA, Motzer R, Voss MH, Hakimi AA. Transcriptomic signatures related to the obesity paradox in patients with clear cell renal cell carcinoma: a cohort study. Lancet Oncol 2019; 21:283-293. [PMID: 31870811 DOI: 10.1016/s1470-2045(19)30797-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Obesity is associated with an increased risk of developing clear cell renal cell carcinoma (RCC) but, paradoxically, obesity is also associated with improved oncological outcomes in this cancer. Because the biological mechanisms underlying this paradoxical association are poorly understood, we aimed to identify transcriptomic differences in primary tumour and peritumoral adipose tissue between obese patients and those at a normal weight. METHODS In this cohort study, we assessed data from five independent clinical cohorts of patients with clear cell RCC aged 18 years and older. Overweight patients were excluded from each cohort for our analysis. We assessed patients from the COMPARZ phase 3 clinical trial, a cohort from the Cancer Genome Atlas (TCGA), and a Memorial Sloan Kettering (MSK) observational immunotherapy cohort for their inclusion into our study. We assessed overall survival in obese patients (those with a body-mass index [BMI] ≥30 kg/m2) and in patients with a normal weight (BMI 18·5-24·9 kg/m2, as per WHO's BMI categories), defined as the time from treatment initiation (in the COMPARZ and MSK immunotherapy cohorts) or surgery (in the TCGA cohort) to the date of any-cause death or of censoring on the day of the last follow-up. We also evaluated and validated transcriptomic differences in the primary tumours of obese patients compared with those of a normal weight. We compared gene-expression differences in peritumoral adipose tissue and tumour tissue in an additional, prospectively collected cohort of patients with non-metastatic clear cell RCC (the MSK peritumoral adipose tissue cohort). We analysed differences in gene expression between obese patients and those at a normal weight in the COMPARZ, TCGA, and peritumoral adipose tissue cohorts. We also assessed the tumour immune microenvironment in a prospective cohort of patients who had nephrectomy for localised RCC at MSK. FINDINGS Of the 453 patients in the COMPARZ trial, 375 (83%) patients had available microarray data, pretreatment BMI measurements, and overall survival data for analyses, and we excluded 119 (26%) overweight patients, leaving a final cohort of 256 (68%) patients from this study for our analyses. From 332 patients in the TCGA cohort, we evaluated clinical and demographic data from 152 (46%) patients with advanced (ie, stages III and IV) clear cell RCC treated by nephrectomy; after exclusion of 59 (39%) overweight patients, our final cohort consisted of 93 (61%) patients. After exclusion of 74 (36%) overweight patients from the initial MSK immunotherapy study population of 203 participants, our final cohort for overall survival analysis comprised 129 (64%) participants. We found that overall survival was longer in obese patients than in those with normal weight in the TCGA cohort, after adjustment for stage or grade (adjusted HR 0·41, 95% CI 0·22-0·75), and in the COMPARZ clinical trial after adjustment for International Metastatic RCC Database (IMDC) risk score (0·68, 0·48-0·96). In the MSK immunotherapy cohort, the inverse association of BMI with mortality (HR 0·54, 95% CI 0·31-0·95) was not significant after adjustment for IMDC risk score (adjusted HR 0·72, 95% CI 0·40-1·30). Tumours of obese patients showed higher angiogenic scores on gene-set enrichment analysis-derived hallmark gene set angiogenesis signatures than did those of patients at a normal weight, but the degree of immune cell infiltration did not differ by BMI. We found increased peritumoral adipose tissue inflammation in obese patients relative to those at a normal weight, especially in peritumoral fat near the tumour. INTERPRETATION We found aspects of the tumour microenvironment that vary by BMI in the tumour and peritumoral adipose tissue, which might contribute to the apparent survival advantage in obese patients with clear cell RCC compared with patients at a normal weight. The complex interplay between the clear cell RCC tumour and peritumoral adipose tissue microenvironment might have clinical relevance and warrants further investigation. FUNDING Ruth L Kirschstein Research Service Award, American Society of Clinical Oncology Young Investigator Award, MSK's Ludwig Center, Weiss Family Kidney Research Fund, Novartis, The Sidney Kimmel Center for Prostate and Urologic Cancers, and the National Institutes of Health (National Cancer Institute) Cancer Center Support Grant.
Collapse
Affiliation(s)
- Alejandro Sanchez
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Surgery, Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helena Furberg
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Immunogenomics & Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynda Vuong
- Immunogenomics & Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasser Ged
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sujata Patil
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Ostrovnaya
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stacey Petruzella
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Roy Mano
- Department of Surgery, Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Coleman
- Department of Surgery, Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Russo
- Department of Surgery, Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine H Liu
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Timothy A Chan
- Immunogenomics & Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
206
|
Paradiso F, Fitzgerald J, Yao S, Barry F, Taraballi F, Gonzalez D, Conlan RS, Francis L. Marine Collagen Substrates for 2D and 3D Ovarian Cancer Cell Systems. Front Bioeng Biotechnol 2019; 7:343. [PMID: 31921795 PMCID: PMC6923181 DOI: 10.3389/fbioe.2019.00343] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022] Open
Abstract
A fundamental structural component of extracellular matrix in all connective and interstitial tissue, collagen is the most abundant protein in the human body. To date, mammalian collagens sources represent the golden standard for multiple biomedical applications, while marine-derived collagens have largely been used in industry (food, pharmaceutical, and cosmetic), with little use in research and clinical applications. Herein we demonstrate the effective use Rhizostoma pulmo jellyfish collagen, a source of biocompatible, sustainable collagen for 2D and 3D cell culture, addressing the global drive for technological developments that result in the replacement of animals and their derived products in research. Jellyfish collagen harbors similar structural features mammalian collagen type I, despite differing slightly in amino acid content. Jellyfish collagen supports ovarian cancer (OvCa) cell line proliferation, cellular morphology and expression of epithelial to mesenchymal transition markers, supporting the use of R. pulmo as a non-mammalian collagen cell culture substrate. Furthermore, R. pulmo collagen is effective in 3D device fabrication such as sponges where it mimics tissue architecture complexity. OvCa cells migrated and differentiated within the R. pulmo collagen 3D scaffolds confirming its suitability for advanced cell culturing applications, providing an excellent alternative to mammalian collagen sources for the culture of human cells.
Collapse
Affiliation(s)
- Francesca Paradiso
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea, United Kingdom.,Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Joan Fitzgerald
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway (NUI), Galway, Ireland
| | - Seydou Yao
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea, United Kingdom
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway (NUI), Galway, Ireland
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Deyarina Gonzalez
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea, United Kingdom
| | - R Steven Conlan
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea, United Kingdom
| |
Collapse
|
207
|
Alkmin S, Brodziski R, Simon H, Hinton D, Goldsmith RH, Patankar M, Campagnola P. Migration dynamics of ovarian epithelial cells on micro-fabricated image-based models of normal and malignant stroma. Acta Biomater 2019; 100:92-104. [PMID: 31568876 DOI: 10.1016/j.actbio.2019.09.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Abstract
A profound remodeling of the collagen in the extracellular matrix (ECM) occurs in human ovarian cancer but it is unknown how this affects migration dynamics and ultimately tumor growth. Here, we investigate the influence of collagen morphology on ovarian cell migration through the use of second harmonic generation (SHG) image-based models of ovarian tumors. The scaffolds are fabricated by multiphoton excited (MPE) polymerization, where the process is akin to 3D printing except it achieves much greater resolution (∼0.5 µm) and utilizes collagen and collagen analogs. We used this technique to create scaffolds with complex 3D submicron features representing the collagen fiber morphology in normal stroma, high risk stroma, benign tumors, and high grade ovarian tumors. We found the highly aligned malignant stromal structure promoted enhanced motility and also increased cell and f-Actin alignment relative to the other tissues. However, using models based on fiber crimping characteristics, we found cells seeded on linear fibers based on normal stromal models yielded the highest degree of alignment but least motility. These results show that both the fiber properties themselves and as well as their overall alignment govern the resulting migration dynamics. These models cannot be synthesized by other conventional fabrication methods and we suggest the MPE image-based fabrication method will enable a variety of studies in cancer biology. STATEMENT OF SIGNIFICANCE: The extracellular matrix collagen in ovarian cancer is highly remodeled but the consequences on cell function remain unknown. It is important to understand the operative cell matrix interactions, as this could lead to better prognostics and better prediction of therapeutic efficacy. We probe migration dynamics using high resolution (∼0.5 µm) multiphoton excited fabrication to synthesize scaffolds whose designs are derived directly from Second Harmonic Generation microscope images of the collagen in normal ovarian tissues as well as benign and malignant tumors. Collectively our results show the importance of the matrix morphology (fiber shape and alignment) on driving cell motility, cell shape and f-Actin alignment. These collagen-based models have complex fiber morphology and cannot be created by conventional fabrication technologies.
Collapse
|
208
|
Abstract
Image analysis in clinical research has evolved at fast pace in the last decade. This review discusses basic concepts ranging from immunohistochemistry to advanced techniques such as multiplex imaging, digital pathology, flow cytometry and intravital microscopy. Tissue imaging
ex vivo is still one of the gold-standards in the field due to feasibility. We describe here different protocols and applications of digital analysis providing basic and clinical researchers with an overview on how to analyse tissue images.
In vivo imaging is not easily accessible to researchers; however, it provides invaluable dynamic information. Overall, we discuss a plethora of techniques that - when combined - constitute a powerful platform for basic and translational cancer research.
Collapse
Affiliation(s)
- Oscar Maiques
- Barts Cancer Institute, John Vane Science Building, Charterhouse Square, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Mirella Georgouli
- Oncology Cell Therapy RU, GlaxoSmithKline, Stevenage, London, SG1 2NY, UK
| | - Victoria Sanz-Moreno
- Barts Cancer Institute, John Vane Science Building, Charterhouse Square, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
209
|
Davis MN, Horne-Badovinac S, Naba A. In-silico definition of the Drosophila melanogaster matrisome. Matrix Biol Plus 2019; 4:100015. [PMID: 33543012 PMCID: PMC7852309 DOI: 10.1016/j.mbplus.2019.100015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 01/02/2023] Open
Abstract
The extracellular matrix (ECM) is an assembly of hundreds of proteins that structurally supports the cells it surrounds and biochemically regulates their functions. Drosophila melanogaster has emerged as a powerful model organism to study fundamental mechanisms underlying ECM protein secretion, ECM assembly, and ECM roles in pathophysiological processes. However, as of today, we do not possess a well-defined list of the components forming the ECM of this organism. We previously reported the development of computational pipelines to define the matrisome - the ensemble of genes encoding ECM and ECM-associated proteins - of humans, mice, zebrafish and C. elegans. Using a similar approach, we report here that our pipeline has identified 641 genes constituting the Drosophila matrisome. We further classify these genes into different structural and functional categories, including an expanded way to classify genes encoding proteins forming apical ECMs. We illustrate how having a comprehensive list of Drosophila matrisome proteins can be used to annotate large proteomic datasets and identify unsuspected roles for the ECM in pathophysiological processes. Last, to aid the dissemination and usage of the proposed definition and categorization of the Drosophila matrisome by the scientific community, our list has been made available through three public portals: The Matrisome Project (http://matrisome.org), The FlyBase (https://flybase.org/), and GLAD (https://www.flyrnai.org/tools/glad/web/).
Collapse
Affiliation(s)
- Martin N. Davis
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| |
Collapse
|
210
|
Graumann J, Finkernagel F, Reinartz S, Stief T, Brödje D, Renz H, Jansen JM, Wagner U, Worzfeld T, Pogge von Strandmann E, Müller R. Multi-platform Affinity Proteomics Identify Proteins Linked to Metastasis and Immune Suppression in Ovarian Cancer Plasma. Front Oncol 2019; 9:1150. [PMID: 31737572 PMCID: PMC6839336 DOI: 10.3389/fonc.2019.01150] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/16/2019] [Indexed: 11/13/2022] Open
Abstract
A central reason behind the poor clinical outcome of patients with high-grade serous carcinoma (HGSC) of the ovary is the difficulty in reliably detecting early occurrence or recurrence of this malignancy. Biomarkers that provide reliable diagnosis of this disease are therefore urgently needed. Systematic proteomic methods that identify HGSC-associated molecules may provide such biomarkers. We applied the antibody-based proximity extension assay (PEA) platform (Olink) for the identification of proteins that are upregulated in the plasma of OC patients. Using binders targeting 368 different plasma proteins, we compared 20 plasma samples from HGSC patients (OC-plasma) with 20 plasma samples from individuals with non-malignant gynecologic disorders (N-plasma). We identified 176 proteins with significantly higher levels in OC-plasma compared to N-plasma by PEA (p < 0.05 by U-test; Benjamini-Hochberg corrected), which are mainly implicated in immune regulation and metastasis-associated processes, such as matrix remodeling, adhesion, migration and proliferation. A number of these proteins have not been reported in previous studies, such as BCAM, CDH6, DDR1, N2DL-2 (ULBP2), SPINT2, and WISP-1 (CCN4). Of these SPINT2, a protease inhibitor mainly derived from tumor cells within the HGSC microenvironment, showed the highest significance (p < 2 × 10−7) similar to the previously described IL-6 and PVRL4 (NECTIN4) proteins. Results were validated by means of the aptamer-based 1.3 k SOMAscan proteomic platform, which revealed a high inter-platform correlation with a median Spearman ρ of 0.62. Likewise, ELISA confirmed the PEA data for 10 out of 12 proteins analyzed, including SPINT2. These findings suggest that in contrast to other entities SPINT2 does not act as a tumor suppressor in HGSC. This is supported by data from the PRECOG and KM-Plotter meta-analysis databases, which point to a tumor-type-specific inverse association of SPINT2 gene expression with survival. Our data also demonstrate that both the PEA and SOMAscan affinity proteomics platforms bear considerable potential for the unbiased discovery of novel disease-associated biomarkers.
Collapse
Affiliation(s)
- Johannes Graumann
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Florian Finkernagel
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Thomas Stief
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps University, Marburg, Germany
| | - Dörte Brödje
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps University, Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM), Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM), Marburg, Germany
| | - Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, Marburg, Germany.,Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Biology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Rolf Müller
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| |
Collapse
|
211
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. FEBS J 2019; 286:4160-4175. [PMID: 31365790 PMCID: PMC6899673 DOI: 10.1111/febs.15028] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/24/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Zuoren Yu
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Jun Mi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Hongqiao International Institute of MedicineTongren HospitalShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
212
|
Mitra S, Tiwari K, Podicheti R, Pandhiri T, Rusch DB, Bonetto A, Zhang C, Mitra AK. Transcriptome Profiling Reveals Matrisome Alteration as a Key Feature of Ovarian Cancer Progression. Cancers (Basel) 2019; 11:cancers11101513. [PMID: 31600962 PMCID: PMC6826756 DOI: 10.3390/cancers11101513] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/30/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecologic malignancy. There is a lack of comprehensive investigation of disease initiation and progression, including gene expression changes during early metastatic colonization. METHODS RNA-sequencing (RNA-seq) was done with matched primary tumors and fallopian tubes (n = 8 pairs) as well as matched metastatic and primary tumors (n = 11 pairs) from ovarian cancer patients. Since these are end point analyses, it was combined with RNA-seq using high-grade serous ovarian cancer cells seeded on an organotypic three-dimensional (3D) culture model of the omentum, mimicking early metastasis. This comprehensive approach revealed key changes in gene expression occurring in ovarian cancer initiation and metastasis, including early metastatic colonization. RESULTS 2987 genes were significantly deregulated in primary tumors compared to fallopian tubes, 845 genes were differentially expressed in metastasis compared to primary tumors and 304 genes were common to both. An assessment of patient metastasis and 3D omental culture model of early metastatic colonization revealed 144 common genes that were altered during early colonization and remain deregulated even in the fully developed metastasis. Deregulation of the matrisome was a key process in early and late metastasis. CONCLUSION These findings will help in understanding the key pathways involved in ovarian cancer progression and eventually targeting those pathways for therapeutic interventions.
Collapse
Affiliation(s)
- Sumegha Mitra
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kartikeya Tiwari
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA.
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA.
| | - Taruni Pandhiri
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA.
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA.
| | - Andrea Bonetto
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Anirban K Mitra
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
213
|
Tripodo C. Time for a "Plan B" in Peritoneal Metastatic Disease. Cancer Res 2019; 79:5-6. [PMID: 30602622 DOI: 10.1158/0008-5472.can-18-3553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/09/2018] [Indexed: 11/16/2022]
Abstract
Peritoneal involvement in cancer is the harbinger of a particularly unfavorable prognosis. The peritoneal cavity microenvironment is skewed toward immunoregulatory conditions promoted by macrophage populations and innate-like B-1 B cells, which provide immune privilege to malignant cell foci. In this issue of Cancer Research, Haro and colleagues demonstrate that triggering innate IgM-mediated B-1a immune responses via pathogen- or danger-associated molecular pattern recognition exerts antitumor effects on peritoneal metastases by inducing classical complement cascade activation. Exploitation of innate B-1 humoral responses and noncellular immunity is a promising strategy to counter the "castling" of metastatic tumor cells in the peritoneal immunoprivileged site.See related article by Haro et al., p. 159.
Collapse
Affiliation(s)
- Claudio Tripodo
- Tumor Immunology Unit, University of Palermo School of Medicine, Palermo, Italy.
| |
Collapse
|
214
|
Diaz Osterman CJ, Ozmadenci D, Kleinschmidt EG, Taylor KN, Barrie AM, Jiang S, Bean LM, Sulzmaier FJ, Jean C, Tancioni I, Anderson K, Uryu S, Cordasco EA, Li J, Chen XL, Fu G, Ojalill M, Rappu P, Heino J, Mark AM, Xu G, Fisch KM, Kolev VN, Weaver DT, Pachter JA, Győrffy B, McHale MT, Connolly DC, Molinolo A, Stupack DG, Schlaepfer DD. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy. eLife 2019; 8:e47327. [PMID: 31478830 PMCID: PMC6721800 DOI: 10.7554/elife.47327] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022] Open
Abstract
Gene copy number alterations, tumor cell stemness, and the development of platinum chemotherapy resistance contribute to high-grade serous ovarian cancer (HGSOC) recurrence. Stem phenotypes involving Wnt-β-catenin, aldehyde dehydrogenase activities, intrinsic platinum resistance, and tumorsphere formation are here associated with spontaneous gains in Kras, Myc and FAK (KMF) genes in a new aggressive murine model of ovarian cancer. Adhesion-independent FAK signaling sustained KMF and human tumorsphere proliferation as well as resistance to cisplatin cytotoxicity. Platinum-resistant tumorspheres can acquire a dependence on FAK for growth. Accordingly, increased FAK tyrosine phosphorylation was observed within HGSOC patient tumors surviving neo-adjuvant chemotherapy. Combining a FAK inhibitor with platinum overcame chemoresistance and triggered cell apoptosis. FAK transcriptomic analyses across knockout and reconstituted cells identified 135 targets, elevated in HGSOC, that were regulated by FAK activity and β-catenin including Myc, pluripotency and DNA repair genes. These studies reveal an oncogenic FAK signaling role supporting chemoresistance.
Collapse
Affiliation(s)
- Carlos J Diaz Osterman
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Duygu Ozmadenci
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Elizabeth G Kleinschmidt
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Kristin N Taylor
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Allison M Barrie
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Shulin Jiang
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Lisa M Bean
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Florian J Sulzmaier
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Christine Jean
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Isabelle Tancioni
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Kristen Anderson
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Sean Uryu
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Edward A Cordasco
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cellular Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cellular Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cellular Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | | | - Pekka Rappu
- Department of BiochemistryUniversity of TurkuTurkuFinland
| | - Jyrki Heino
- Department of BiochemistryUniversity of TurkuTurkuFinland
| | - Adam M Mark
- Department of MedicineUCSD Center for Computational Biology & BioinformaticsLa JollaUnited States
| | - Guorong Xu
- Department of MedicineUCSD Center for Computational Biology & BioinformaticsLa JollaUnited States
| | - Kathleen M Fisch
- Department of MedicineUCSD Center for Computational Biology & BioinformaticsLa JollaUnited States
| | | | | | | | - Balázs Győrffy
- Institute of EnzymologyHungarian Academy of SciencesBudapestHungary
- 2nd Department of PediatricsSemmelweis UniversityBudapestHungary
| | - Michael T McHale
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | | | - Alfredo Molinolo
- Department of PathologyMoores UCSD Cancer CenterLa JollaUnited States
| | - Dwayne G Stupack
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| | - David D Schlaepfer
- Department of Obstetrics, Gynecology and Reproductive SciencesMoores UCSD Cancer CenterLa JollaUnited States
| |
Collapse
|
215
|
Exploring the extracellular matrix in health and disease using proteomics. Essays Biochem 2019; 63:417-432. [DOI: 10.1042/ebc20190001] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Abstract
The extracellular matrix (ECM) is a complex assembly of hundreds of proteins that constitutes the scaffold of multicellular organisms. In addition to providing architectural and mechanical support to the surrounding cells, it conveys biochemical signals that regulate cellular processes including proliferation and survival, fate determination, and cell migration. Defects in ECM protein assembly, decreased ECM protein production or, on the contrary, excessive ECM accumulation, have been linked to many pathologies including cardiovascular and skeletal diseases, cancers, and fibrosis. The ECM thus represents a potential reservoir of prognostic biomarkers and therapeutic targets. However, our understanding of the global protein composition of the ECM and how it changes during pathological processes has remained limited until recently.
In this mini-review, we provide an overview of the latest methodological advances in sample preparation and mass spectrometry-based proteomics that have permitted the profiling of the ECM of now dozens of normal and diseased tissues, including tumors and fibrotic lesions.
Collapse
|
216
|
Martinez VG, Park D, Acton SE. Immunotherapy: breaching the barriers for cancer treatment. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180214. [PMID: 31431180 PMCID: PMC6627023 DOI: 10.1098/rstb.2018.0214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2019] [Indexed: 12/18/2022] Open
Abstract
The great ambition to treat cancer through harnessing a patient's own immune responses has started to become reality. Clinical trials have shown impressive results and some patients reaching the end of existing treatment options have achieved full remission. Yet the response rate even within the most promising trials remain at just 30-40% of patients. To date, the focus of immunotherapy research has been to identify tumour antigens, and to enhance activation of effector lymphocytes. Yet this is only the first step to effective immunotherapy for a broader range of patients. Activated cytotoxic T cells can only act on their tumour cell targets if they have free and easy access to all tumour regions. Solid tumours are complex, heterogeneous environments which vary greatly in their physical properties. We must now focus our efforts on understanding how factors such as the composition, density and geometry of tumour extracellular matrix acts to impede or promote immune cell infiltration and activation, and work to design novel pharmacological interventions which restore and enhance leucocyte trafficking within solid tumours. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Victor G. Martinez
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Danielle Park
- Tumour Cell Biology Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
217
|
Gordon S, Plüddemann A. The Mononuclear Phagocytic System. Generation of Diversity. Front Immunol 2019; 10:1893. [PMID: 31447860 PMCID: PMC6696592 DOI: 10.3389/fimmu.2019.01893] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023] Open
Abstract
We are living through an unprecedented accumulation of data on gene expression by macrophages, reflecting their origin, distribution, and localization within all organs of the body. While the extensive heterogeneity of the cells of the mononuclear phagocyte system is evident, the functional significance of their diversity remains incomplete, nor is the mechanism of diversification understood. In this essay we review some of the implications of what we know, and draw attention to issues to be clarified in further research, taking advantage of the powerful genetic, cellular, and molecular tools now available. Our thesis is that macrophage specialization and functions go far beyond immunobiology, while remaining an essential contributor to innate as well as adaptive immunity.
Collapse
Affiliation(s)
- Siamon Gordon
- College of Medicine, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Annette Plüddemann
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
218
|
Mitra AK, Yang-Hartwich Y. Tumor microenvironment and immunology of ovarian cancer: 12th Biennial Rivkin Center Ovarian Cancer Research Symposium. Int J Gynecol Cancer 2019; 29:s12-s15. [PMID: 31462543 DOI: 10.1136/ijgc-2019-000666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 11/03/2022] Open
Abstract
The 12th Biennial Ovarian Cancer Research Symposium organized by the Rivkin Center for Ovarian Cancer and the American Association for Cancer Research held on September 13-15, 2018 covered cutting edge and relevant research topics in ovarian cancer biology and therapy. Sessions included detection and prevention, genomics and molecular mechanisms, tumor microenvironment and immunology, novel therapeutics, and an education session. In this article we provide an overview of the key findings presented in the tumor microenvironment and immunology session.
Collapse
Affiliation(s)
- Anirban K Mitra
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Yang Yang-Hartwich
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
219
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. THE FEBS JOURNAL 2019. [PMID: 31365790 DOI: 10.1111/febs.15028.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Zuoren Yu
- Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Jun Mi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
220
|
Burgos-Panadero R, Lucantoni F, Gamero-Sandemetrio E, Cruz-Merino LDL, Álvaro T, Noguera R. The tumour microenvironment as an integrated framework to understand cancer biology. Cancer Lett 2019; 461:112-122. [PMID: 31325528 DOI: 10.1016/j.canlet.2019.07.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 01/18/2023]
Abstract
Cancer cells all share the feature of being immersed in a complex environment with altered cell-cell/cell-extracellular element communication, physicochemical information, and tissue functions. The so-called tumour microenvironment (TME) is becoming recognised as a key factor in the genesis, progression and treatment of cancer lesions. Beyond genetic mutations, the existence of a malignant microenvironment forms the basis for a new perspective in cancer biology where connections at the system level are fundamental. From this standpoint, different aspects of tumour lesions such as morphology, aggressiveness, prognosis and treatment response can be considered under an integrated vision, giving rise to a new field of study and clinical management. Nowadays, somatic mutation theory is complemented with study of TME components such as the extracellular matrix, immune compartment, stromal cells, metabolism and biophysical forces. In this review we examine recent studies in this area and complement them with our own research data to propose a classification of stromal changes. Exploring these avenues and gaining insight into malignant phenotype remodelling, could reveal better ways to characterize this disease and its potential treatment.
Collapse
Affiliation(s)
- Rebeca Burgos-Panadero
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Federico Lucantoni
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain
| | - Esther Gamero-Sandemetrio
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | | | - Tomás Álvaro
- CIBERONC, Madrid, Spain; Hospital Verge de la Cinta, Tortosa, Tarragona, Spain.
| | - Rosa Noguera
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain.
| |
Collapse
|
221
|
Bast RC, Matulonis UA, Sood AK, Ahmed AA, Amobi AE, Balkwill FR, Wielgos-Bonvallet M, Bowtell DDL, Brenton JD, Brugge JS, Coleman RL, Draetta GF, Doberstein K, Drapkin RI, Eckert MA, Edwards RP, Elias KM, Ennis D, Futreal A, Gershenson DM, Greenberg RA, Huntsman DG, Ji JXY, Kohn EC, Iavarone C, Lengyel ER, Levine DA, Lord CJ, Lu Z, Mills GB, Modugno F, Nelson BH, Odunsi K, Pilsworth JA, Rottapel RK, Powell DJ, Shen L, Shih LM, Spriggs DR, Walton J, Zhang K, Zhang R, Zou L. Critical questions in ovarian cancer research and treatment: Report of an American Association for Cancer Research Special Conference. Cancer 2019; 125:1963-1972. [PMID: 30835824 PMCID: PMC6557260 DOI: 10.1002/cncr.32004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022]
Abstract
Substantial progress has been made in understanding ovarian cancer at the molecular and cellular level. Significant improvement in 5-year survival has been achieved through cytoreductive surgery, combination platinum-based chemotherapy, and more effective treatment of recurrent cancer, and there are now more than 280,000 ovarian cancer survivors in the United States. Despite these advances, long-term survival in late-stage disease has improved little over the last 4 decades. Poor outcomes relate, in part, to late stage at initial diagnosis, intrinsic drug resistance, and the persistence of dormant drug-resistant cancer cells after primary surgery and chemotherapy. Our ability to accelerate progress in the clinic will depend on the ability to answer several critical questions regarding this disease. To assess current answers, an American Association for Cancer Research Special Conference on "Critical Questions in Ovarian Cancer Research and Treatment" was held in Pittsburgh, Pennsylvania, on October 1-3, 2017. Although clinical, translational, and basic investigators conducted much of the discussion, advocates participated in the meeting, and many presentations were directly relevant to patient care, including treatment with poly adenosine diphosphate ribose polymerase (PARP) inhibitors, attempts to improve immunotherapy by overcoming the immune suppressive effects of the microenvironment, and a better understanding of the heterogeneity of the disease.
Collapse
Affiliation(s)
- Robert C. Bast
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Anil K. Sood
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | - Zhen Lu
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Brad H. Nelson
- University of British Columbia, Canada
- BC Cancer Agency, Canada
| | | | | | | | | | - Li Shen
- Roswell Park Cancer Institute, Buffalo, NY
| | - le-Ming Shih
- Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | | - Lee Zou
- Massachusetts General Hospital, Boston, MD
| |
Collapse
|
222
|
Matera DL, Wang WY, Smith MR, Shikanov A, Baker BM. Fiber Density Modulates Cell Spreading in 3D Interstitial Matrix Mimetics. ACS Biomater Sci Eng 2019; 5:2965-2975. [PMID: 33405599 DOI: 10.1021/acsbiomaterials.9b00141] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular phenotype is heavily influenced by the extracellular matrix (ECM), a complex and tissue-specific three-dimensional structure with distinct biophysical and biochemical properties. As naturally derived cell culture platforms are difficult to controllably modulate, engineered synthetic ECMs have facilitated our understanding of how specific matrix properties direct cell behavior. However, synthetic approaches typically lack fibrous topography, a hallmark of stromal and interstitial ECMs in vivo. To construct tunable biomimetic models with physiologic microstructure, we developed a versatile approach to generate modular fibrous architectures in 3D. Photo-cross-linkable polymers were electrospun, photopatterned into desired lengths, and coencapsulated alongside cells within natural biopolymer, semisynthetic, and synthetic hydrogels. Cells encapsulated within fiber-reinforced hydrogel composites (FHCs) demonstrated accelerated spreading rates compared to in gels lacking such fibrous topography. Furthermore, increases in fiber density at constant bulk hydrogel elastic modulus produced morphologically distinct cell populations and modulated cellular mechanosensing in 3D, as evidenced by increased nuclear localization of the mechanosensitive transcription factor, Yes-associated protein (YAP). This work documents the impact of physical guidance cues in 3D and establishes a novel approach to generating more physiologic tissue- and disease-specific biomimetic models.
Collapse
|
223
|
Yin S, Du J, Zhang J, Zhang X, Ma K. Identification of Key Genes and Pathway for Ovarian Neoplasms Using the OVDM1 Cell Line Based on Bioinformatics Analysis. Med Sci Monit 2019; 25:4305-4313. [PMID: 31177266 PMCID: PMC6582691 DOI: 10.12659/msm.915422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Ovarian neoplasms are the fifth most common cancer affecting the health of women, and they are the most lethal gynecologic malignancies; however, the etiology of ovarian neoplasms remains largely unknown. There is an urgent need to further broaden the understanding of the development mechanism of ovarian neoplasms through in vitro research using different cell lines. Material/Methods To screen the differentially expressed genes (DEGs) that may play critical roles in OVDM1 (an ovarian cancer cell line), the public microarray data (GSE70264) were downloaded and screened for DEGs. Then, Gene Ontology (GO) function analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed. To screen hub genes, the protein–protein interaction network was constructed. The expression level and survival analysis of hub genes in patients with ovarian neoplasms were also analyzed. Results There were 79 upregulated and 926 downregulated DEGs detected, and the biological processes of the GO analysis were enriched in extracellular matrix organization, extracellular structure organization, and chromosome segregation, whereas, the KEGG pathway analysis was enriched in cell cycle and cell adhesion molecules. The hub gene BIRC5, which might play a key role in ovarian neoplasms, was further screened. Conclusions The present study could deepen the understanding of the molecular mechanism of ovarian neoplasms using the OVDM1 cell line, which could be useful in developing clinical treatments of ovarian neoplasms.
Collapse
Affiliation(s)
- Songna Yin
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Juan Du
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Jie Zhang
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Xiang Zhang
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Ke Ma
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|
224
|
Pan-Cancer analysis of the expression and regulation of matrisome genes across 32 tumor types. Matrix Biol Plus 2019; 1:100004. [PMID: 33543003 PMCID: PMC7852311 DOI: 10.1016/j.mbplus.2019.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/28/2022] Open
Abstract
The microenvironment plays a central role in cancer, and neoplastic cells actively shape it to their needs by complex arrays of extracellular matrix (ECM) proteins, enzymes, cytokines and growth factors collectively referred to as the matrisome. Studies on the cancer matrisome have been performed for single or few neoplasms, but a more systematic analysis is still missing. Here we present a Pan-Cancer study of matrisome gene expression in 10,487 patients across 32 tumor types, supplemented with transcription factors (TFs) and driver genes/pathways regulating each tumor's matrisome. We report on 919 TF-target pairs, either used specifically or shared across tumor types, and their prognostic significance, 40 master regulators, 31 overarching regulatory pathways and the potential for druggability with FDA-approved cancer drugs. These results provide a comprehensive transcriptional architecture of the cancer matrisome and suggest the need for development of specific matrisome-targeting approaches for future therapies. In-depth characterization of matrisome gene expression and regulation in 10,487 patients across 32 human tumor types. Identification of transcription factor (TF) and “master regulators” governing each cancer’s matrisome. Analysis unveils therapeutic possibilities and suggests new treatments by repurposing of FDA-approved cancer drugs.
Collapse
|
225
|
Di Modugno F, Colosi C, Trono P, Antonacci G, Ruocco G, Nisticò P. 3D models in the new era of immune oncology: focus on T cells, CAF and ECM. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:117. [PMID: 30898166 PMCID: PMC6429763 DOI: 10.1186/s13046-019-1086-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitor therapy has changed clinical practice for patients with different cancers, since these agents have demonstrated a significant improvement of overall survival and are effective in many patients. However, an intrinsic or acquired resistance frequently occur and biomarkers predictive of responsiveness should help in patient selection and in defining the adequate treatment options. A deep analysis of the complexity of the tumor microenvironment is likely to further advance the field and hopefully identify more effective combined immunotherapeutic strategies. Here we review the current knowledge on tumor microenvironment, focusing on T cells, cancer associated fibroblasts and extracellular matrix. The use of 3D cell culture models to resemble tumor microenvironment landscape and to screen immunomodulatory drugs is also reviewed.
Collapse
Affiliation(s)
- Francesca Di Modugno
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy.
| | - Cristina Colosi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Paola Trono
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy
| | - Giuseppe Antonacci
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Paola Nisticò
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
226
|
Natarajan S, Foreman KM, Soriano MI, Rossen NS, Shehade H, Fregoso DR, Eggold JT, Krishnan V, Dorigo O, Krieg AJ, Heilshorn SC, Sinha S, Fuh KC, Rankin EB. Collagen Remodeling in the Hypoxic Tumor-Mesothelial Niche Promotes Ovarian Cancer Metastasis. Cancer Res 2019; 79:2271-2284. [PMID: 30862717 DOI: 10.1158/0008-5472.can-18-2616] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/27/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
Peritoneal metastases are the leading cause of morbidity and mortality in high-grade serous ovarian cancer (HGSOC). Accumulating evidence suggests that mesothelial cells are an important component of the metastatic microenvironment in HGSOC. However, the mechanisms by which mesothelial cells promote metastasis are unclear. Here, we report that the HGSOC tumor-mesothelial niche was hypoxic, and hypoxic signaling enhanced collagen I deposition by mesothelial cells. Specifically, hypoxic signaling increased expression of lysyl oxidase (LOX) in mesothelial and ovarian cancer cells to promote collagen crosslinking and tumor cell invasion. The mesothelial niche was enriched with fibrillar collagen in human and murine omental metastases. Pharmacologic inhibition of LOX reduced tumor burden and collagen remodeling in murine omental metastases. These findings highlight an important role for hypoxia and mesothelial cells in the modification of the extracellular matrix and tumor invasion in HGSOC. SIGNIFICANCE: This study identifies HIF/LOX signaling as a potential therapeutic target to inhibit collagen remodeling and tumor progression in HGSOC.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2271/F1.large.jpg.
Collapse
Affiliation(s)
- Suchitra Natarajan
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Kaitlyn M Foreman
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Michaela I Soriano
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Ninna S Rossen
- Department of Radiation Oncology, Stanford University, Palo Alto, California.,Department of Materials Science and Engineering, Stanford University, Palo Alto, California
| | - Hussein Shehade
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Daniel R Fregoso
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Joshua T Eggold
- Department of Radiation Oncology, Stanford University, Palo Alto, California
| | - Venkatesh Krishnan
- Department of Obstetrics and Gynecology, Stanford University, Palo Alto, California
| | - Oliver Dorigo
- Department of Obstetrics and Gynecology, Stanford University, Palo Alto, California
| | - Adam J Krieg
- Oregon Health & Science University, Portland, Oregon
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Palo Alto, California
| | | | - Katherine C Fuh
- Division of Gynecologic Oncology, Washington University, St. Louis, Missouri
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University, Palo Alto, California. .,Department of Obstetrics and Gynecology, Stanford University, Palo Alto, California
| |
Collapse
|
227
|
Holenstein CN, Horvath A, Schär B, Schoenenberger AD, Bollhalder M, Goedecke N, Bartalena G, Otto O, Herbig M, Guck J, Müller DA, Snedeker JG, Silvan U. The relationship between metastatic potential and in vitro mechanical properties of osteosarcoma cells. Mol Biol Cell 2019; 30:887-898. [PMID: 30785850 PMCID: PMC6589788 DOI: 10.1091/mbc.e18-08-0545] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma is the most frequent primary tumor of bone and is characterized by its high tendency to metastasize in lungs. Although treatment in cases of early diagnosis results in a 5-yr survival rate of nearly 60%, the prognosis for patients with secondary lesions at diagnosis is poor, and their 5-yr survival rate remains below 30%. In the present work, we have used a number of analytical methods to investigate the impact of increased metastatic potential on the biophysical properties and force generation of osteosarcoma cells. With that aim, we used two paired osteosarcoma cell lines, with each one comprising a parental line with low metastatic potential and its experimentally selected, highly metastatic form. Mechanical characterization was performed by means of atomic force microscopy, tensile biaxial deformation, and real-time deformability, and cell traction was measured using two-dimensional and micropost-based traction force microscopy. Our results reveal that the low metastatic osteosarcoma cells display larger spreading sizes and generate higher forces than the experimentally selected, highly malignant variants. In turn, the outcome of cell stiffness measurements strongly depends on the method used and the state of the probed cell, indicating that only a set of phenotyping methods provides the full picture of cell mechanics.
Collapse
Affiliation(s)
- Claude N Holenstein
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Aron Horvath
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Barbara Schär
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Angelina D Schoenenberger
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Maja Bollhalder
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Nils Goedecke
- Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Guido Bartalena
- Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Oliver Otto
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany.,Zentrum für Innovationskompetenz, Universität Greifswald, 17489 Greifswald, Germany
| | - Maik Herbig
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Daniel A Müller
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland
| | - Jess G Snedeker
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| | - Unai Silvan
- Biomechanics Laboratory, University Hospital Balgrist, University of Zürich, 8008 Zürich, Switzerland.,Institute for Biomechanics, ETH Zurich, 8008 Zürich, Switzerland
| |
Collapse
|
228
|
Lee W, Ko SY, Mohamed MS, Kenny HA, Lengyel E, Naora H. Neutrophils facilitate ovarian cancer premetastatic niche formation in the omentum. J Exp Med 2019; 216:176-194. [PMID: 30567719 PMCID: PMC6314534 DOI: 10.1084/jem.20181170] [Citation(s) in RCA: 322] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/09/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer preferentially metastasizes to the omentum, a fatty tissue characterized by immune structures called milky spots, but the cellular dynamics that direct this tropism are unknown. Here, we identified that neutrophil influx into the omentum is a prerequisite premetastatic step in orthotopic ovarian cancer models. Ovarian tumor-derived inflammatory factors stimulated neutrophils to mobilize and extrude chromatin webs called neutrophil extracellular traps (NETs). NETs were detected in the omentum of ovarian tumor-bearing mice before metastasis and of women with early-stage ovarian cancer. NETs, in turn, bound ovarian cancer cells and promoted metastasis. Omental metastasis was decreased in mice with neutrophil-specific deficiency of peptidylarginine deiminase 4 (PAD4), an enzyme that is essential for NET formation. Blockade of NET formation using a PAD4 pharmacologic inhibitor also decreased omental colonization. Our findings implicate NET formation in rendering the premetastatic omental niche conducive for implantation of ovarian cancer cells and raise the possibility that blockade of NET formation prevents omental metastasis.
Collapse
Affiliation(s)
- WonJae Lee
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Song Yi Ko
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Muhaned S Mohamed
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hilary A Kenny
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL
| | - Ernst Lengyel
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL
| | - Honami Naora
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
229
|
An evolving story of the metastatic voyage of ovarian cancer cells: cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene 2018; 38:2885-2898. [PMID: 30568223 PMCID: PMC6755962 DOI: 10.1038/s41388-018-0637-x] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
Metastasis is a complex multistep process that involves critical interactions between cancer cells and a variety of stromal components in the tumor microenvironment, which profoundly influence the different aspects of the metastatic cascade and organ tropism of disseminating cancer cells. Ovarian cancer is the most lethal gynecological malignancy and is characterized by peritoneal disseminated metastasis. Evidence has demonstrated that ovarian cancer possesses specific metastatic tropism for the adipose-rich omentum, which has a pivotal role in the creation of the metastatic tumor microenvironment in the intraperitoneal cavity. Considering the distinct biology of ovarian cancer metastasis, the elucidation of the cellular and molecular mechanisms underlying the reciprocal interplay between ovarian cancer cells and surrounding stromal cell types in the adipose-rich metastatic microenvironment will provide further insights into the development of novel therapeutic approaches for patients with advanced ovarian cancer. Herein, we review the biological mechanisms that regulate the highly orchestrated crosstalk between ovarian cancer cells and various cancer-associated stromal cells in the metastatic tumor microenvironment with regard to the omentum by illustrating how different stromal cells concertedly contribute to the development of ovarian cancer metastasis and metastatic tropism for the omentum.
Collapse
|
230
|
Loessner D, Rockstroh A, Shokoohmand A, Holzapfel BM, Wagner F, Baldwin J, Boxberg M, Schmalfeldt B, Lengyel E, Clements JA, Hutmacher DW. A 3D tumor microenvironment regulates cell proliferation, peritoneal growth and expression patterns. Biomaterials 2018; 190-191:63-75. [PMID: 30396040 DOI: 10.1016/j.biomaterials.2018.10.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/09/2018] [Accepted: 10/14/2018] [Indexed: 02/07/2023]
Abstract
Peritoneal invasion through the mesothelial cell layer is a hallmark of ovarian cancer metastasis. Using tissue engineering technologies, we recreated an ovarian tumor microenvironment replicating this aspect of disease progression. Ovarian cancer cell-laden hydrogels were combined with mesothelial cell-layered melt electrospun written scaffolds and characterized with proliferation and transcriptomic analyses and used as intraperitoneal xenografts. Here we show increased cancer cell proliferation in these 3D co-cultures, which we validated using patient-derived cells and linked to peritoneal tumor growth in vivo. Transcriptome-wide expression analysis identified IGFBP7, PTGS2, VEGFC and FGF2 as bidirectional factors deregulated in 3D co-cultures compared to 3D mono-cultures, which we confirmed by immunohistochemistry of xenograft and patient-derived tumor tissues and correlated with overall and progression-free survival. These factors were further increased upon expression of kallikrein-related proteases. This clinically predictive model allows us to mimic the complexity and processes of the metastatic disease that may lead to therapies that protect from peritoneal invasion or delay the development of metastasis.
Collapse
Affiliation(s)
- Daniela Loessner
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Anja Rockstroh
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Ali Shokoohmand
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Boris M Holzapfel
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany
| | - Ferdinand Wagner
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 4, 80337 Munich, Germany
| | - Jeremy Baldwin
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Melanie Boxberg
- Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Barbara Schmalfeldt
- Gynecologic Department, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, 5841 South Maryland Avenue, MC2050, Chicago, IL 60637, USA
| | - Judith A Clements
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Dietmar W Hutmacher
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332-0405, USA; Institute for Advanced Study, Technical University Munich, Lichtenbergstr. 2a, 85748 Garching, Germany.
| |
Collapse
|
231
|
Cunnea P, Gorgy T, Petkos K, Gowers SAN, Lu H, Morera C, Wu W, Lawton P, Nixon K, Leong CL, Sorbi F, Domenici L, Paterson A, Curry E, Gabra H, Boutelle MG, Drakakis EM, Fotopoulou C. Clinical value of bioelectrical properties of cancerous tissue in advanced epithelial ovarian cancer patients. Sci Rep 2018; 8:14695. [PMID: 30279418 PMCID: PMC6168525 DOI: 10.1038/s41598-018-32720-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/11/2018] [Indexed: 01/20/2023] Open
Abstract
Currently, there are no valid pre-operatively established biomarkers or algorithms that can accurately predict surgical and clinical outcome for patients with advanced epithelial ovarian cancer (EOC). In this study, we suggest that profiling of tumour parameters such as bioelectrical-potential and metabolites, detectable by electronic sensors, could facilitate the future development of devices to better monitor disease and predict surgical and treatment outcomes. Biopotential was recorded, using a potentiometric measurement system, in ex vivo paired non-cancerous and cancerous omental tissues from advanced stage EOC (n = 36), and lysates collected for metabolite measurement by microdialysis. Consistently different biopotential values were detected in cancerous tissue versus non-cancerous tissue across all cases (p < 0.001). High tumour biopotential levels correlated with advanced tumour stage (p = 0.048) and tumour load, and negatively correlated with stroma. Within our EOC cohort and specifically the high-grade serous subtype, low biopotential levels associated with poorer progression-free survival (p = 0.0179, p = 0.0143 respectively). Changes in biopotential levels significantly correlated with common apoptosis related pathways. Lactate and glucose levels measured in paired tissues showed significantly higher lactate/glucose ratio in tissues with low biopotential (p < 0.01, n = 12). Our study proposes the feasibility of biopotential and metabolite monitoring as a biomarker modality profiling EOC to predict surgical and clinical outcomes.
Collapse
Affiliation(s)
- Paula Cunnea
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Tommy Gorgy
- Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | - Haonan Lu
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Cristina Morera
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Wen Wu
- Department of Bioengineering, Imperial College, London, UK
| | - Phillip Lawton
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Katherine Nixon
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Chi Leng Leong
- Department of Bioengineering, Imperial College, London, UK
| | - Flavia Sorbi
- Department of Surgery and Cancer, Imperial College, London, UK
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Lavinia Domenici
- Department of Surgery and Cancer, Imperial College, London, UK
- Department of Obstetrics, Gynecology and Urologic Sciences, University "Sapienza" of Rome, Rome, Italy
| | - Andrew Paterson
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Ed Curry
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Hani Gabra
- Department of Surgery and Cancer, Imperial College, London, UK
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | | | | |
Collapse
|
232
|
Yang M, McKay D, Pollard JW, Lewis CE. Diverse Functions of Macrophages in Different Tumor Microenvironments. Cancer Res 2018; 78:5492-5503. [PMID: 30206177 PMCID: PMC6171744 DOI: 10.1158/0008-5472.can-18-1367] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/21/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Tumor-associated macrophages are a major constituent of malignant tumors and are known to stimulate key steps in tumor progression. In our review in this journal in 2006, we postulated that functionally distinct subsets of these cells exist in different areas within solid tumors. Here, we review the many experimental and clinical studies conducted since then to investigate the function(s), regulation, and clinical significance of macrophages in these sites. The latter include three sites of cancer cell invasion, tumor nests, the tumor stroma, and areas close to, or distant from, the tumor vasculature. A more complete understanding of macrophage diversity in tumors could lead to the development of more selective therapies to restore the formidable, anticancer functions of these cells. Cancer Res; 78(19); 5492-503. ©2018 AACR.
Collapse
Affiliation(s)
- Ming Yang
- Department of Oncology & Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Daniel McKay
- Department of Oncology & Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Claire E Lewis
- Department of Oncology & Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom.
| |
Collapse
|
233
|
Gupta V, Yull F, Khabele D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018. [PMID: 30274280 DOI: 10.3390/cancers10100366] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Fiona Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA.
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| |
Collapse
|
234
|
Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018; 10:cancers10100366. [PMID: 30274280 PMCID: PMC6210537 DOI: 10.3390/cancers10100366] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/22/2018] [Accepted: 09/23/2018] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
|
235
|
Gupta V, Yull F, Khabele D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018. [PMID: 30274280 DOI: 10.3390/cancers10100366]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Fiona Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA.
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| |
Collapse
|
236
|
Hudson LG, Gillette JM, Kang H, Rivera MR, Wandinger-Ness A. Ovarian Tumor Microenvironment Signaling: Convergence on the Rac1 GTPase. Cancers (Basel) 2018; 10:cancers10100358. [PMID: 30261690 PMCID: PMC6211091 DOI: 10.3390/cancers10100358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment for epithelial ovarian cancer is complex and rich in bioactive molecules that modulate cell-cell interactions and stimulate numerous signal transduction cascades. These signals ultimately modulate all aspects of tumor behavior including progression, metastasis and therapeutic response. Many of the signaling pathways converge on the small GTPase Ras-related C3 botulinum toxin substrate (Rac)1. In addition to regulating actin cytoskeleton remodeling necessary for tumor cell adhesion, migration and invasion, Rac1 through its downstream effectors, regulates cancer cell survival, tumor angiogenesis, phenotypic plasticity, quiescence, and resistance to therapeutics. In this review we discuss evidence for Rac1 activation within the ovarian tumor microenvironment, mechanisms of Rac1 dysregulation as they apply to ovarian cancer, and the potential benefits of targeting aberrant Rac1 activity in this disease. The potential for Rac1 contribution to extraperitoneal dissemination of ovarian cancer is addressed.
Collapse
Affiliation(s)
- Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Jennifer M Gillette
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Huining Kang
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Melanie R Rivera
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Angela Wandinger-Ness
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
237
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018; 10:E302. [PMID: 30200478 PMCID: PMC6162424 DOI: 10.3390/cancers10090302] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
238
|
Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
|
239
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
240
|
Muir A, Danai LV, Vander Heiden MG. Microenvironmental regulation of cancer cell metabolism: implications for experimental design and translational studies. Dis Model Mech 2018; 11:dmm035758. [PMID: 30104199 PMCID: PMC6124553 DOI: 10.1242/dmm.035758] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cancers have an altered metabolism, and there is interest in understanding precisely how oncogenic transformation alters cellular metabolism and how these metabolic alterations can translate into therapeutic opportunities. Researchers are developing increasingly powerful experimental techniques to study cellular metabolism, and these techniques have allowed for the analysis of cancer cell metabolism, both in tumors and in ex vivo cancer models. These analyses show that, while factors intrinsic to cancer cells such as oncogenic mutations, alter cellular metabolism, cell-extrinsic microenvironmental factors also substantially contribute to the metabolic phenotype of cancer cells. These findings highlight that microenvironmental factors within the tumor, such as nutrient availability, physical properties of the extracellular matrix, and interactions with stromal cells, can influence the metabolic phenotype of cancer cells and might ultimately dictate the response to metabolically targeted therapies. In an effort to better understand and target cancer metabolism, this Review focuses on the experimental evidence that microenvironmental factors regulate tumor metabolism, and on the implications of these findings for choosing appropriate model systems and experimental approaches.
Collapse
Affiliation(s)
- Alexander Muir
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura V Danai
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
241
|
Chitty JL, Filipe EC, Lucas MC, Herrmann D, Cox TR, Timpson P. Recent advances in understanding the complexities of metastasis. F1000Res 2018; 7. [PMID: 30135716 DOI: 10.12688/f1000research.15064.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Tumour metastasis is a dynamic and systemic process. It is no longer seen as a tumour cell-autonomous program but as a multifaceted and complex series of events, which is influenced by the intrinsic cellular mutational burden of cancer cells and the numerous bidirectional interactions between malignant and non-malignant cells and fine-tuned by the various extrinsic cues of the extracellular matrix. In cancer biology, metastasis as a process is one of the most technically challenging aspects of cancer biology to study. As a result, new platforms and technologies are continually being developed to better understand this process. In this review, we discuss some of the recent advances in metastasis and how the information gleaned is re-shaping our understanding of metastatic dissemination.
Collapse
Affiliation(s)
- Jessica L Chitty
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Elysse C Filipe
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Morghan C Lucas
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| |
Collapse
|
242
|
The Tumor Microenvironment Takes Center Stage in Ovarian Cancer Metastasis. Trends Cancer 2018; 4:517-519. [DOI: 10.1016/j.trecan.2018.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/07/2018] [Indexed: 11/20/2022]
|
243
|
Chitty JL, Filipe EC, Lucas MC, Herrmann D, Cox TR, Timpson P. Recent advances in understanding the complexities of metastasis. F1000Res 2018; 7. [PMID: 30135716 PMCID: PMC6073095 DOI: 10.12688/f1000research.15064.2] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Tumour metastasis is a dynamic and systemic process. It is no longer seen as a tumour cell-autonomous program but as a multifaceted and complex series of events, which is influenced by the intrinsic cellular mutational burden of cancer cells and the numerous bidirectional interactions between malignant and non-malignant cells and fine-tuned by the various extrinsic cues of the extracellular matrix. In cancer biology, metastasis as a process is one of the most technically challenging aspects of cancer biology to study. As a result, new platforms and technologies are continually being developed to better understand this process. In this review, we discuss some of the recent advances in metastasis and how the information gleaned is re-shaping our understanding of metastatic dissemination.
Collapse
Affiliation(s)
- Jessica L Chitty
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Elysse C Filipe
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Morghan C Lucas
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| |
Collapse
|
244
|
Socovich AM, Naba A. The cancer matrisome: From comprehensive characterization to biomarker discovery. Semin Cell Dev Biol 2018; 89:157-166. [PMID: 29964200 DOI: 10.1016/j.semcdb.2018.06.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 02/07/2023]
Abstract
Tumor progression and dissemination critically depend on support from the tumor microenvironment, the ensemble of cellular and acellular components surrounding and interacting with tumor cells. The extracellular matrix (ECM), the complex scaffolding of hundreds of proteins organizing cells in tissues, is a major component of the tumor microenvironment. It orchestrates cellular processes including proliferation, migration, and invasion, that are highly dysregulated during cancer progression. Alterations in ECM abundance, integrity, and mechanical properties have been correlated with poorer prognosis for cancer patients. Yet the ECM proteome, or "matrisome," of tumors remained until recently largely unexplored. This review will present the recent developments in computational and proteomic technologies that have allowed the comprehensive characterization of the ECM of different tumor types and microenvironmental niches. These approaches have resulted in the definition of protein signatures distinguishing tumors from normal tissues, tumors of different stages, primary from secondary tumors, and tumors from other diseased states such as fibrosis. Moreover, recent studies have demonstrated that the levels of expression of certain genes encoding ECM and ECM-associated proteins is prognostic of cancer patient survival and can thus serve as biomarkers. Last, proteomic studies have permitted the identification of novel ECM proteins playing functional roles in cancer progression. Such proteins have the potential to be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Alexandra M Socovich
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
245
|
Mushtaq MU, Papadas A, Pagenkopf A, Flietner E, Morrow Z, Chaudhary SG, Asimakopoulos F. Tumor matrix remodeling and novel immunotherapies: the promise of matrix-derived immune biomarkers. J Immunother Cancer 2018; 6:65. [PMID: 29970158 PMCID: PMC6029413 DOI: 10.1186/s40425-018-0376-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023] Open
Abstract
Recent advances in our understanding of the dynamics of cellular cross-talk have highlighted the significance of host-versus-tumor effect that can be harnessed with immune therapies. Tumors exploit immune checkpoints to evade adaptive immune responses. Cancer immunotherapy has witnessed a revolution in the past decade with the development of immune checkpoint inhibitors (ICIs), monoclonal antibodies against cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or their ligands, such as PD1 ligand 1 (PD-L1). ICIs have been reported to have activity against a broad range of tumor types, in both solid organ and hematologic malignancy contexts. However, less than one-third of the patients achieve a durable and meaningful treatment response. Expression of immune checkpoint ligands (e.g., PD-L1), mutational burden and tumor-infiltrating lymphocytes are currently used as biomarkers for predicting response to ICIs. However, they do not reliably predict which patients will benefit from these therapies. There is dire need to discover novel biomarkers to predict treatment efficacy and to identify areas for development of combination strategies to improve response rates. Emerging evidence suggests key roles of tumor extracellular matrix (ECM) components and their proteolytic remodeling products in regulating each step of the cancer-immunity cycle. Here we review tumor matrix dynamics and matrix remodeling in context of anti-tumor immune responses and immunotherapy and propose the exploration of matrix-based biomarkers to identify candidates for immune therapy.
Collapse
Affiliation(s)
- Muhammad Umair Mushtaq
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Athanasios Papadas
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Adam Pagenkopf
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Evan Flietner
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Zachary Morrow
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Sibgha Gull Chaudhary
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Fotis Asimakopoulos
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA.
| |
Collapse
|
246
|
Mohammadi H, Sahai E. Mechanisms and impact of altered tumour mechanics. Nat Cell Biol 2018; 20:766-774. [PMID: 29950570 DOI: 10.1038/s41556-018-0131-2] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
Abstract
The physical characteristics of tumours are intricately linked to the tumour phenotype and difficulties during treatment. Many factors contribute to the increased stiffness of tumours; from increased matrix deposition, matrix remodelling by forces from cancer cells and stromal fibroblasts, matrix crosslinking, increased cellularity, and the build-up of both solid and interstitial pressure. Increased stiffness then feeds back to increase tumour invasiveness and reduce therapy efficacy. Increased understanding of this interplay is offering new therapeutic avenues.
Collapse
|
247
|
Cai Q, Fan Q, Buechlein A, Miller D, Nephew KP, Liu S, Wan J, Xu Y. Changes in mRNA/protein expression and signaling pathways in in vivo passaged mouse ovarian cancer cells. PLoS One 2018; 13:e0197404. [PMID: 29927933 PMCID: PMC6013233 DOI: 10.1371/journal.pone.0197404] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/01/2018] [Indexed: 01/03/2023] Open
Abstract
The cure rate for late stage epithelial ovarian cancer (EOC) has not significantly improved over several decades. New and more effective targets and treatment modalities are urgently needed. RNA-seq analyses of a syngeneic EOC cell pair, representing more and less aggressive tumor cells in vivo were conducted. Bioinformatics analyses of the RNA-seq data and biological signaling and function studies have identified new targets, such as ZIP4 in EOC. Many up-regulated tumor promoting signaling pathways have been identified which are mainly grouped into three cellular activities: 1) cell proliferation and apoptosis resistance; 2) cell skeleton and adhesion changes; and 3) carbohydrate metabolic reprograming. Unexpectedly, lipid metabolism has been the major down-regulated signaling pathway in the more aggressive EOC cells. In addition, we found that hypoxic responsive genes were at the center stage of regulation and detected functional changes were related to cancer stem cell-like activities. Moreover, our genetic, cellular, biochemical, and lipidomic analyses indicated that cells grown in 2D vs. 3D, or attached vs. suspended had dramatic changes. The important clinical implications of peritoneal cavity floating tumor cells are supported by the data proved in this work. Overall, the RNA-seq data provide a landscape of gene expression alterations during tumor progression.
Collapse
Affiliation(s)
- Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Aaron Buechlein
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - David Miller
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - Kenneth P. Nephew
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - Sheng Liu
- Collaborative Core for Cancer Bioinformatics (C3B), Indiana University Simon Cancer Center, Indianapolis, Indiana, United States of America
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jun Wan
- Collaborative Core for Cancer Bioinformatics (C3B), Indiana University Simon Cancer Center, Indianapolis, Indiana, United States of America
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
248
|
Shafi AA, Schiewer MJ, de Leeuw R, Dylgjeri E, McCue PA, Shah N, Gomella LG, Lallas CD, Trabulsi EJ, Centenera MM, Hickey TE, Butler LM, Raj G, Tilley WD, Cukierman E, Knudsen KE. Patient-derived Models Reveal Impact of the Tumor Microenvironment on Therapeutic Response. Eur Urol Oncol 2018; 1:325-337. [PMID: 30467556 DOI: 10.1016/j.euo.2018.04.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Androgen deprivation therapy is a first-line treatment for disseminated prostate cancer (PCa). However, virtually all tumors become resistant and recur as castration-resistant PCa, which has no durable cure. One major hurdle in the development of more effective therapies is the lack of preclinical models that adequately recapitulate the heterogeneity of PCa, significantly hindering the ability to accurately predict therapeutic response. Objective To leverage the ex vivo culture method termed patient-derived explant (PDE) to examine the impact of PCa therapeutics on a patient-by-patient basis. Design setting and participants Fresh PCa tissue from patients who underwent radical prostatectomy was cultured as PDEs to examine therapeutic response. Outcome measurements and statistical analysis The impact of genomic and chemical perturbations in PDEs was assessed using various parameters (eg, AR levels, Ki67 staining, and desmoplastic indices). Results and limitations PDE maintained the integrity of the native tumor microenvironment (TME), tumor tissue morphology, viability, and endogenous hormone signaling. Tumor cells in this model system exhibited de novo proliferative capacity. Examination of the native TME in the PDE revealed a first-in-field insight into patient-specific desmoplastic stromal indices and predicted responsiveness to AR-directed therapeutics. Conclusions The PDE model allows for a comprehensive evaluation of individual tumors in their native TME to ultimately develop more effective therapeutic regimens tailored to individuals. Discernment of novel stromal markers may provide a basis for applying precision medicine in treating advanced PCa, which would have a transformative effect on patient outcomes. Patient summary In this study, an innovative model system was used to more effectively mimic human disease. The patient-derived explant (PDE) system can be used to predict therapeutic response and identify novel targets in advanced disease. Thus, the PDE will be an asset for the development of novel metrics for the implementation of precision medicine in prostate cancer.The patient-derived explant (PDE) model allows for a comprehensive evaluation of individual human tumors in their native tumor microenvironment (TME). TME analysis revealed first-in-field insight into predicted tumor responsiveness to AR-directed therapeutics through evaluation of patient-specific desmoplastic stromal indices.
Collapse
Affiliation(s)
- Ayesha A Shafi
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthew J Schiewer
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Renée de Leeuw
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emanuela Dylgjeri
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Neelima Shah
- Cancer Biology, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Leonard G Gomella
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Costas D Lallas
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edouard J Trabulsi
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Margaret M Centenera
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Prostate Cancer Research Centre and Freemason's Foundation Centre for Men's Health, School of Medicine, University of Adelaide, Adelaide, Australia.,South Australian Health and Medician Research Institute, Adelaide, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Prostate Cancer Research Centre and Freemason's Foundation Centre for Men's Health, School of Medicine, University of Adelaide, Adelaide, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Prostate Cancer Research Centre and Freemason's Foundation Centre for Men's Health, School of Medicine, University of Adelaide, Adelaide, Australia.,South Australian Health and Medician Research Institute, Adelaide, Australia
| | - Ganesh Raj
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Prostate Cancer Research Centre and Freemason's Foundation Centre for Men's Health, School of Medicine, University of Adelaide, Adelaide, Australia
| | - Edna Cukierman
- Cancer Biology, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Departments of Cancer Biology and Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
249
|
Antibodies and methods for immunohistochemistry of extracellular matrix proteins. Matrix Biol 2018; 71-72:10-27. [PMID: 29730502 DOI: 10.1016/j.matbio.2018.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/12/2023]
Abstract
The diversity of extracellular matrix (ECM) proteins encoded in mammalian genomes and detected by proteomic analyses generates a need for well validated antibodies against these proteins. We present characterization of a large number of antibodies against ECM proteins, from both commercial and academic sources, together with discussion of methods and strategies for their effective use in immunohistochemistry and illustrations of their efficacy. These data should be of value to investigators seeking well validated antibodies to ECM proteins of interest and save significant time and money tracking down effective reagents.
Collapse
|
250
|
Filipe EC, Chitty JL, Cox TR. Charting the unexplored extracellular matrix in cancer. Int J Exp Pathol 2018; 99:58-76. [PMID: 29671911 DOI: 10.1111/iep.12269] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is present in all solid tissues and considered a master regulator of cell behaviour and phenotype. The importance of maintaining the correct biochemical and biophysical properties of the ECM, and the subsequent regulation of cell and tissue homeostasis, is illustrated by the simple fact that the ECM is highly dysregulated in many different types of disease, especially cancer. The loss of tissue ECM homeostasis and integrity is seen as one of the hallmarks of cancer and typically defines transitional events in progression and metastasis. The vast majority of cancer studies place an emphasis on exploring the behaviour and intrinsic signalling pathways of tumour cells. Their goal was to identify ways to target intracellular pathways regulating cancer. Cancer progression and metastasis are powerfully influenced by the ECM and thus present a vast, unexplored repository of anticancer targets that we are only just beginning to tap into. Deconstructing the complexity of the tumour ECM landscape and identifying the interactions between the many cell types, soluble factors and extracellular-matrix proteins have proved challenging. Here, we discuss some of the emerging tools and platforms being used to catalogue and chart the ECM in cancer.
Collapse
Affiliation(s)
- Elysse C Filipe
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Jessica L Chitty
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Thomas R Cox
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|