1
|
Ruff SM, Pawlik TM. A Review of Translational Research for Targeted Therapy for Metastatic Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15051395. [PMID: 36900187 PMCID: PMC10000872 DOI: 10.3390/cancers15051395] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Colorectal cancer is the third most common cause of cancer-related death in the United States, with 20% of patients presenting with metastatic disease at the time of diagnosis. Metastatic colon cancer is often treated with a combination of surgery, systemic therapy (chemotherapy, biologic therapy, immunotherapy), and/or regional therapy (hepatic artery infusion pumps). Utilizing the molecular and pathologic features of the primary tumor to tailor treatment for patients may improve overall survival. Rather than a "one size fits all" approach, a more nuanced treatment plan guided by the unique features of a patient's tumor and the tumor's microenvironment can more effectively treat the disease. Basic science work to elucidate new drug targets, understand mechanisms of evasion, and develop drugs and drug combinations is critical to inform clinical trials and identify novel, effective therapies for metastatic colorectal cancer. Through the lens of key targets for metastatic colorectal cancer, this review discusses how work in the basic science lab translates into clinical trials.
Collapse
|
2
|
Quadri M, Marconi A, Sandhu SK, Kiss A, Efimova T, Palazzo E. Investigating Cutaneous Squamous Cell Carcinoma in vitro and in vivo: Novel 3D Tools and Animal Models. Front Med (Lausanne) 2022; 9:875517. [PMID: 35646967 PMCID: PMC9131878 DOI: 10.3389/fmed.2022.875517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/19/2022] [Indexed: 12/07/2022] Open
Abstract
Cutaneous Squamous Cell Carcinoma (cSCC) represents the second most common type of skin cancer, which incidence is continuously increasing worldwide. Given its high frequency, cSCC represents a major public health problem. Therefore, to provide the best patients’ care, it is necessary having a detailed understanding of the molecular processes underlying cSCC development, progression, and invasion. Extensive efforts have been made in developing new models allowing to study the molecular pathogenesis of solid tumors, including cSCC tumors. Traditionally, in vitro studies were performed with cells grown in a two-dimensional context, which, however, does not represent the complexity of tumor in vivo. In the recent years, new in vitro models have been developed aiming to mimic the three-dimensionality (3D) of the tumor, allowing the evaluation of tumor cell-cell and tumor-microenvironment interaction in an in vivo-like setting. These models include spheroids, organotypic cultures, skin reconstructs and organoids. Although 3D models demonstrate high potential to enhance the overall knowledge in cancer research, they lack systemic components which may be solved only by using animal models. Zebrafish is emerging as an alternative xenotransplant model in cancer research, offering a high-throughput approach for drug screening and real-time in vivo imaging to study cell invasion. Moreover, several categories of mouse models were developed for pre-clinical purpose, including xeno- and syngeneic transplantation models, autochthonous models of chemically or UV-induced skin squamous carcinogenesis, and genetically engineered mouse models (GEMMs) of cSCC. These models have been instrumental in examining the molecular mechanisms of cSCC and drug response in an in vivo setting. The present review proposes an overview of in vitro, particularly 3D, and in vivo models and their application in cutaneous SCC research.
Collapse
Affiliation(s)
- Marika Quadri
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Marconi
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Simran K Sandhu
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Alexi Kiss
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Tatiana Efimova
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Elisabetta Palazzo
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
3
|
Singh D, Deosarkar SP, Cadogan E, Flemington V, Bray A, Zhang J, Reiserer RS, Schaffer DK, Gerken GB, Britt CM, Werner EM, Gibbons FD, Kostrzewski T, Chambers CE, Davies EJ, Montoya AR, Fok JHL, Hughes D, Fabre K, Wagoner MP, Wikswo JP, Scott CW. A microfluidic system that replicates pharmacokinetic (PK) profiles in vitro improves prediction of in vivo efficacy in preclinical models. PLoS Biol 2022; 20:e3001624. [PMID: 35617197 PMCID: PMC9135222 DOI: 10.1371/journal.pbio.3001624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 04/11/2022] [Indexed: 11/19/2022] Open
Abstract
Test compounds used on in vitro model systems are conventionally delivered to cell culture wells as fixed concentration bolus doses; however, this poorly replicates the pharmacokinetic (PK) concentration changes seen in vivo and reduces the predictive value of the data. Herein, proof-of-concept experiments were performed using a novel microfluidic device, the Microformulator, which allows in vivo like PK profiles to be applied to cells cultured in microtiter plates and facilitates the investigation of the impact of PK on biological responses. We demonstrate the utility of the device in its ability to reproduce in vivo PK profiles of different oncology compounds over multiweek experiments, both as monotherapy and drug combinations, comparing the effects on tumour cell efficacy in vitro with efficacy seen in in vivo xenograft models. In the first example, an ERK1/2 inhibitor was tested using fixed bolus dosing and Microformulator-replicated PK profiles, in 2 cell lines with different in vivo sensitivities. The Microformulator-replicated PK profiles were able to discriminate between cell line sensitivities, unlike the conventional fixed bolus dosing. In a second study, murine in vivo PK profiles of multiple Poly(ADP-Ribose) Polymerase 1/2 (PARP) and DNA-dependent protein kinase (DNA-PK) inhibitor combinations were replicated in a FaDu cell line resulting in a reduction in cell growth in vitro with similar rank ordering to the in vivo xenograft model. Additional PK/efficacy insight into theoretical changes to drug exposure profiles was gained by using the Microformulator to expose FaDu cells to the DNA-PK inhibitor for different target coverage levels and periods of time. We demonstrate that the Microformulator enables incorporating PK exposures into cellular assays to improve in vitro-in vivo translation understanding for early therapeutic insight.
Collapse
Affiliation(s)
| | - Sudhir P. Deosarkar
- Oncology Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, United States of America
| | - Elaine Cadogan
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Vikki Flemington
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alysha Bray
- CN Bio Innovations Limited, Cambridge, United Kingdom
| | - Jingwen Zhang
- Bioscience, Oncology R&D, AstraZeneca, Boston, Massachusetts, United States of America
| | - Ronald S. Reiserer
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
| | - David K. Schaffer
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
| | - Gregory B. Gerken
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
| | - Clayton M. Britt
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
| | - Erik M. Werner
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
| | - Francis D. Gibbons
- DMPK, Oncology R&D, AstraZeneca, Boston, Massachusetts, United States of America
| | | | | | - Emma J. Davies
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - David Hughes
- CN Bio Innovations Limited, Cambridge, United Kingdom
| | - Kristin Fabre
- MPS Center of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, United States of America
| | - Matthew P. Wagoner
- Oncology Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, United States of America
| | - John P. Wikswo
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Nashville, Tennessee, United States of America
- Departments of Biomedical Engineering and Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Clay W. Scott
- Oncology Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, United States of America
| |
Collapse
|
4
|
Ballke S, Heid I, Mogler C, Braren R, Schwaiger M, Weichert W, Steiger K. Correlation of in vivo imaging to morphomolecular pathology in translational research: challenge accepted. EJNMMI Res 2021; 11:83. [PMID: 34453623 PMCID: PMC8401369 DOI: 10.1186/s13550-021-00826-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/15/2021] [Indexed: 12/26/2022] Open
Abstract
Correlation of in vivo imaging to histomorphological pathology in animal models requires comparative interdisciplinary expertise of different fields of medicine. From the morphological point of view, there is an urgent need to improve histopathological evaluation in animal model-based research to expedite translation into clinical applications. While different other fields of translational science were standardized over the last years, little was done to improve the pipeline of experimental pathology to ensure reproducibility based on pathological expertise in experimental animal models with respect to defined guidelines and classifications. Additionally, longitudinal analyses of preclinical models often use a variety of imaging methods and much more attention should be drawn to enable for proper co-registration of in vivo imaging methods with the ex vivo morphological read-outs. Here we present the development of the Comparative Experimental Pathology (CEP) unit embedded in the Institute of Pathology of the Technical University of Munich during the Collaborative Research Center 824 (CRC824) funding period together with selected approaches of histomorphological techniques for correlation of in vivo imaging to morphomolecular pathology.
Collapse
Affiliation(s)
- Simone Ballke
- School of Medicine, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Irina Heid
- School of Medicine, Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carolin Mogler
- School of Medicine, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Rickmer Braren
- School of Medicine, Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Markus Schwaiger
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wilko Weichert
- School of Medicine, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- School of Medicine, Institute of Pathology, Technical University of Munich, Munich, Germany.
| |
Collapse
|
5
|
Skok K, Gradišnik L, Maver U, Kozar N, Sobočan M, Takač I, Arko D, Kavalar R. Gynaecological cancers and their cell lines. J Cell Mol Med 2021; 25:3680-3698. [PMID: 33650759 PMCID: PMC8051715 DOI: 10.1111/jcmm.16397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022] Open
Abstract
Cell lines are widely used for various research purposes including cancer and drug research. Recently, there have been studies that pointed to discrepancies in the literature and usage of cell lines. That is why we have prepared a comprehensive overview of the most common gynaecological cancer cell lines, their literature, a list of currently available cell lines, and new findings compared with the original studies. A literature review was conducted via MEDLINE, PubMed and ScienceDirect for reviews in the last 5 years to identify research and other studies related to gynaecological cancer cell lines. We present an overview of the current literature with reference to the original studies and pointed to certain inconsistencies in the literature. The adherence to culturing rulesets and the international guidelines helps in minimizing replication failure between institutions. Evidence from the latest research suggests that despite certain drawbacks, variations of cancer cell lines can also be useful in regard to a more diverse genomic landscape.
Collapse
Affiliation(s)
- Kristijan Skok
- Department of pathology, General Hospital Graz II, Graz, Austria.,Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Lidija Gradišnik
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Nejc Kozar
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Monika Sobočan
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Iztok Takač
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Darja Arko
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rajko Kavalar
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Department of Pathology, University Medical Center Maribor, Maribor, Slovenia
| |
Collapse
|
6
|
Markov SD, Gonzalez D, Mehla K. Preclinical Models for Studying the Impact of Macrophages on Cancer Cachexia. CURRENT PROTOCOLS IN PHARMACOLOGY 2020; 91:e80. [PMID: 33264501 PMCID: PMC8099022 DOI: 10.1002/cpph.80] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer-associated cachexia is defined by loss of weight and muscle mass, and by the potential loss of adipose tissue accompanied by insulin resistance and increased resting energy expenditure. Cachexia is most prevalent in pancreatic cancer, the third leading cause of cancer-related deaths. While various factors interact to induce cachexia, the precise mechanisms underlying this clinical condition are not fully understood. Clinically relevant animal models of cachexia are needed given the lack of standard diagnostic methods or treatments for this condition. Described in this article are in vitro and in vivo models used to study the role of macrophages in the induction of cachexia in pancreatic cancer. Included are procedures for isolating and culturing bone marrow-derived macrophages, harvesting tumor- and macrophage-derived conditioned medium, and studying the effect of conditioned medium on C2C12 myotubes. Also described are procedures involving the use of an orthotopic model of pancreatic cancer, including a method for examining skeletal muscle atrophy in this model. © 2020 Wiley Periodicals LLC. Basic Protocol 1: In vitro model of pancreatic tumor-induced cachexia using C2C12 cell lines (myotube model) Support Protocol 1: Molecular evaluation of cachectic markers in C2C12 myotubes using real-time PCR and immunoblotting Basic Protocol 2: In vivo model to study cachectic phenotype in pancreatic tumor-bearing mice Support Protocol 2: Evaluation of cachectic markers in the skeletal muscle of tumor-bearing mice.
Collapse
Affiliation(s)
- Spas Dimitrov Markov
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Daisy Gonzalez
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kamiya Mehla
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
7
|
Porter RJ, Murray GI, McLean MH. Current concepts in tumour-derived organoids. Br J Cancer 2020; 123:1209-1218. [PMID: 32728094 PMCID: PMC7555542 DOI: 10.1038/s41416-020-0993-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/28/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer comprises a collection of highly proliferative and heterogeneous cells growing within an adaptive and evolving tumour microenvironment. Cancer survival rates have significantly improved following decades of cancer research. However, many experimental and preclinical studies do not translate to the bedside, reflecting the challenges of modelling the complexities and multicellular basis of human disease. Organoids are novel, complex, three-dimensional ex vivo tissue cultures that are derived from embryonic stem cells, induced pluripotent stem cells or tissue-resident progenitor cells, and represent a near-physiological model for studying cancer. Organoids develop by self-organisation, and can accurately represent the diverse genetic, cellular and pathophysiological hallmarks of cancer. In addition, co-culture methods and the ability to genetically manipulate these organoids have widened their utility in cancer research. Organoids thus offer a new and exciting platform for studying cancer and directing personalised therapies. This review aims to highlight how organoids are shaping the future of cancer research.
Collapse
Affiliation(s)
- Ross J Porter
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | - Graeme I Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK
| | - Mairi H McLean
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK.
| |
Collapse
|
8
|
Zanoni M, Cortesi M, Zamagni A, Arienti C, Pignatta S, Tesei A. Modeling neoplastic disease with spheroids and organoids. J Hematol Oncol 2020; 13:97. [PMID: 32677979 PMCID: PMC7364537 DOI: 10.1186/s13045-020-00931-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer is a complex disease in which both genetic defects and microenvironmental components contribute to the development, progression, and metastasization of disease, representing major hurdles in the identification of more effective and safer treatment regimens for patients. Three-dimensional (3D) models are changing the paradigm of preclinical cancer research as they more closely resemble the complex tissue environment and architecture found in clinical tumors than in bidimensional (2D) cell cultures. Among 3D models, spheroids and organoids represent the most versatile and promising models in that they are capable of recapitulating the heterogeneity and pathophysiology of human cancers and of filling the gap between conventional 2D in vitro testing and animal models. Such 3D systems represent a powerful tool for studying cancer biology, enabling us to model the dynamic evolution of neoplastic disease from the early stages to metastatic dissemination and the interactions with the microenvironment. Spheroids and organoids have recently been used in the field of drug discovery and personalized medicine. The combined use of 3D models could potentially improve the robustness and reliability of preclinical research data, reducing the need for animal testing and favoring their transition to clinical practice. In this review, we summarize the recent advances in the use of these 3D systems for cancer modeling, focusing on their innovative translational applications, looking at future challenges, and comparing them with most widely used animal models.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Michela Cortesi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alice Zamagni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Arienti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Pignatta
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
9
|
Patient-derived model systems and the development of next-generation anticancer therapeutics. Curr Opin Chem Biol 2020; 56:72-78. [DOI: 10.1016/j.cbpa.2020.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
10
|
Wang D, Zhang H, Fang X, Cao D, Liu H. Pan-cancer analysis reveals the role of long non-coding RNA LINC01614 as a highly cancer-dependent oncogene and biomarker. Oncol Lett 2020; 20:1383-1399. [PMID: 32724381 PMCID: PMC7377058 DOI: 10.3892/ol.2020.11648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
Long intergenic non-coding RNA 1614 (LINC01614) is highly expressed in several malignant tumor types, suggesting that it may act as an oncogene. However, the specific roles of LINC01614 in malignant tumors have remained elusive. To examine the expression pattern of LINC01614 in various malignancies, a comprehensive pan-cancer analysis was performed using public databases, including 53 normal tissue types and 32 cancer datasets with samples from 9,091 patients. The results were validated using reverse transcription-quantitative PCR analysis of tissue specimens from patients. LINC01614 expression was upregulated in most malignant tumors, thus demonstrating diagnostic potential. Furthermore, upregulation of LINC01614 was associated with poor overall survival in the majority of cases. However, the association with clinical outcome was highly cancer-dependent; LINC01614 appeared to be an oncogene and diagnostic/prognostic biomarker in cancers of the digestive, respiratory, nervous and endocrine systems, as well as breast and head and neck cancer, but not in the cancers of the reproductive system or some of the urinary system. High LINC01614 expression was also markedly associated with the epithelial-mesenchymal transition (EMT) and associated signaling pathways. Overall, the present results suggest that LINC01614 is an EMT-associated oncogene that influences the metastasis and prognosis of several cancers, thus highlighting its potential as a novel diagnostic and prognostic marker.
Collapse
Affiliation(s)
- Dingding Wang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Hong Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xiaolian Fang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, P.R. China
| | - Dingfang Cao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
11
|
Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, Chan TA, Coukos G, Demaria S, Deutsch E, Draganov D, Edelson RL, Formenti SC, Fucikova J, Gabriele L, Gaipl US, Gameiro SR, Garg AD, Golden E, Han J, Harrington KJ, Hemminki A, Hodge JW, Hossain DMS, Illidge T, Karin M, Kaufman HL, Kepp O, Kroemer G, Lasarte JJ, Loi S, Lotze MT, Manic G, Merghoub T, Melcher AA, Mossman KL, Prosper F, Rekdal Ø, Rescigno M, Riganti C, Sistigu A, Smyth MJ, Spisek R, Stagg J, Strauss BE, Tang D, Tatsuno K, van Gool SW, Vandenabeele P, Yamazaki T, Zamarin D, Zitvogel L, Cesano A, Marincola FM. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer 2020; 8:e000337. [PMID: 32209603 PMCID: PMC7064135 DOI: 10.1136/jitc-2019-000337] [Citation(s) in RCA: 663] [Impact Index Per Article: 132.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York City, New York, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Université de Paris, Paris, France
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Sarah Warren
- NanoString Technologies, Seattle, Washington, USA
| | - Sandy Adjemian
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Cancer Biology, KU Leuevn, Leuven, Belgium
| | - Aitziber Buqué Martinez
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - George Coukos
- Ludwig Institute for Cancer Research and Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, New York, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM "Molecular Radiotherapy and therapeutic innovation", U1030 Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- SIRIC SOCRATES, DHU Torino, Faculté de Medecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Richard L Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jitka Fucikova
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Udo S Gaipl
- Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jian Han
- iRepertoire, Inc, Huntsville, Alabama, USA
| | - Kevin J Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital/Institute of Cancer Research National Institute for Health Biomedical Research Centre, London, UK
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Tim Illidge
- University of Manchester, NIHR Manchester Biomedical Research Centre, Christie Hospital, Manchester, UK
| | - Michael Karin
- Department of Pharmacology and Pathology, University of California at San Diego (UCSD) School of Medicine, La Jolla, California, USA
| | - Howard L Kaufman
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Replimune, Inc, Woburn, Massachusetts, USA
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Guido Kroemer
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1138, Paris, France
- Sorbonne Université, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Juan Jose Lasarte
- Program of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gwenola Manic
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, MSKCC, New York City, New York, USA
- Weill Cornell Medical College, New York City, New York, USA
- Parker Institute for Cancer Immunotherapy, MSKCC, New York City, New York, USA
| | | | | | - Felipe Prosper
- Hematology and Cell Therapy, Clinica Universidad de Navarra, Pamplona, Spain
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
- Interdepartmental Research Center of Molecular Biotechnology, University of Torino, Torino, Italy
| | - Antonella Sistigu
- UOSD Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Radek Spisek
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec City, Canada
- Institut du Cancer de Montréal, Montréal, Quebec City, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Quebec City, Canada
| | - Bryan E Strauss
- Centro de Investigação Translacional em Oncologia/LIM24, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
- Methusalem program, Ghent University, Ghent, Belgium
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Dmitriy Zamarin
- Department of Medicine, Weill Cornell Medical College, New York City, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe labellisée par la Ligue contre le cancer, Gustave Roussy, Villejuif, France
- Faculty of Medicine, University of Paris Sud/Paris Saclay, Le Kremlin-Bicêtre, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | | | | |
Collapse
|
12
|
Abstract
Endometrial cancer is one of the most common gynaecological malignancies worldwide. One type of research in this field is the growing of cell lines (CLs) and cultures, which can be used to explore the biological mechanisms of cancer. The purpose of this review is to offer an overview of the current literature and highlight the importance of correct CL studies. We carried out a literature analysis of more than 60 articles from the Pubmed, Medline databases that were almost exclusively published in indexed journals in the last 10 years as well as the primary originating scientific studies of specific CLs. We then summarized the newest findings and recommendations. Cell lines are becoming widely used as in vitro tumour models. Recent work has shown inconsistencies in nomenclature and culturing of CLs. Their genomic evolution leads to a high degree of variation across CL strains therefore it is of the utmost importance to recognize the variability within laboratory cancer models. Laboratories must adapt, incorporate additional characterisation techniques and view this situation as an opportunity to improve the reproducibility of pre-clinical cancer research. The authors offer a comprehensive literature review about endometrial cancer CLs, a review of the current literature and advice on culturing CLs.
Collapse
|
13
|
Tomasin R, Martin ACBM, Cominetti MR. Metastasis and cachexia: alongside in clinics, but not so in animal models. J Cachexia Sarcopenia Muscle 2019; 10:1183-1194. [PMID: 31436396 PMCID: PMC6903449 DOI: 10.1002/jcsm.12475] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/06/2019] [Accepted: 06/12/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer cachexia is a paraneoplastic syndrome characterized by lean mass wasting (with or without fat mass decrease), culminating in involuntary weight loss, which is the key clinical observation nowadays. There is a notable lack of studies involving animal models to mimic the clinical reality, which are mostly patients with cachexia and metastatic disease. This mismatch between the clinical reality and animal models could at least partly contribute to the poor translation observed in the field. In this paper, we retrieved and compared animal models used for cachexia research from 2017 and 10 years earlier (2007) and observed that very little has changed. Especially, clinically relevant models where cachexia is studied in an orthotopic or metastatic context were and still are very scarce. Finally, we described and supported the biological rationale behind why, despite technical challenges, these two phenomena-metastasis and cachexia-should be modelled in parallel, highlighting the overlapping pathways between them. To sum up, this review aims to contribute to rethinking and possibly switching the models currently used for cachexia research, to hopefully obtain better and more translational outcomes.
Collapse
Affiliation(s)
- Rebeka Tomasin
- Laboratory of Biology of Aging (LABEN), Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
| | | | - Márcia Regina Cominetti
- Laboratory of Biology of Aging (LABEN), Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
14
|
Steiger K, Ballke S, Yen HY, Seelbach O, Alkhamas A, Boxberg M, Schwamborn K, Knolle PA, Weichert W, Mogler C. [Histopathological research laboratories in translational research : Conception and integration into the infrastructure of pathological institutes]. DER PATHOLOGE 2019; 40:172-178. [PMID: 30027333 DOI: 10.1007/s00292-018-0458-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A systematic review of histopathology from experimental animal systems is an essential part of up-to-date biomedical research. Pathologists at university hospitals are especially and increasingly challenged by these specialized and time-consuming duties. This article presents and analyzes a new laboratory structure of comparative experimental pathology-jointly lead by veterinary and human pathologists-which might solve this problem. The focus is on the establishment and full integration of this laboratory structure into a local, regional, and nationwide biomedical research cluster. A detailed comparison with an established structure of routine histopathology laboratories discusses merits and benefits as well as disadvantages.
Collapse
Affiliation(s)
- K Steiger
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland. .,Comparative Experimental Pathology, Technische Universität München, München, Deutschland. .,Vergleichende Experimentelle Pathologie, Institut für Pathologie, Technische Universität München, Trogerstraße 18, 81675, München, Deutschland.
| | - S Ballke
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland
| | - H-Y Yen
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland.,Partnerstandort München, Deutsches Konsortium für Translationale Krebsforschung, München, Deutschland
| | - O Seelbach
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland
| | - A Alkhamas
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland.,Partnerstandort München, Deutsches Konsortium für Translationale Krebsforschung, München, Deutschland
| | - M Boxberg
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland
| | - K Schwamborn
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland
| | - P A Knolle
- Institut für Molekulare Immunologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - W Weichert
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland.,Partnerstandort München, Deutsches Konsortium für Translationale Krebsforschung, München, Deutschland
| | - C Mogler
- Institut für allgemeine Pathologie und pathologische Anatomie, Technische Universität München, München, Deutschland.,Comparative Experimental Pathology, Technische Universität München, München, Deutschland
| |
Collapse
|
15
|
Hooker SE, Woods-Burnham L, Bathina M, Lloyd S, Gorjala P, Mitra R, Nonn L, Kimbro KS, Kittles RA. Genetic Ancestry Analysis Reveals Misclassification of Commonly Used Cancer Cell Lines. Cancer Epidemiol Biomarkers Prev 2019; 28:1003-1009. [PMID: 30787054 PMCID: PMC6548687 DOI: 10.1158/1055-9965.epi-18-1132] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/20/2018] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Given the scarcity of cell lines from underrepresented populations, it is imperative that genetic ancestry for these cell lines is characterized. Consequences of cell line mischaracterization include squandered resources and publication retractions. METHODS We calculated genetic ancestry proportions for 15 cell lines to assess the accuracy of previous race/ethnicity classification and determine previously unknown estimates. DNA was extracted from cell lines and genotyped for ancestry informative markers representing West African (WA), Native American (NA), and European (EUR) ancestry. RESULTS Of the cell lines tested, all previously classified as White/Caucasian were accurately described with mean EUR ancestry proportions of 97%. Cell lines previously classified as Black/African American were not always accurately described. For instance, the 22Rv1 prostate cancer cell line was recently found to carry mixed genetic ancestry using a much smaller panel of markers. However, our more comprehensive analysis determined the 22Rv1 cell line carries 99% EUR ancestry. Most notably, the E006AA-hT prostate cancer cell line, classified as African American, was found to carry 92% EUR ancestry. We also determined the MDA-MB-468 breast cancer cell line carries 23% NA ancestry, suggesting possible Afro-Hispanic/Latina ancestry. CONCLUSIONS Our results suggest predominantly EUR ancestry for the White/Caucasian-designated cell lines, yet high variance in ancestry for the Black/African American-designated cell lines. In addition, we revealed an extreme misclassification of the E006AA-hT cell line. IMPACT Genetic ancestry estimates offer more sophisticated characterization leading to better contextualization of findings. Ancestry estimates should be provided for all cell lines to avoid erroneous conclusions in disparities literature.
Collapse
Affiliation(s)
- Stanley E Hooker
- Division of Health Equities, Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Leanne Woods-Burnham
- Division of Health Equities, Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Madhavi Bathina
- Division of Health Equities, Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Stacy Lloyd
- Department of Molecular and Cellular Biology and Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Priyatham Gorjala
- College of Medicine, Roseman University of Health Sciences, Las Vegas, Nevada
| | - Ranjana Mitra
- College of Medicine, Roseman University of Health Sciences, Las Vegas, Nevada
| | - Larisa Nonn
- The Department of Pathology, University of Illinois, Chicago, Illinois
| | - K Sean Kimbro
- Biomedical/Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, North Carolina
| | - Rick A Kittles
- Division of Health Equities, Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
16
|
Black PC, Goebell PJ, Kamat AM, Nawroth R, Seiler R, Williams SB, Schmitz-Dräger BJ. Editorial: Basic research in bladder cancer - refining the tools. 3rd IBCN seminars series 1. Urol Oncol 2019; 38:855-857. [PMID: 30683454 DOI: 10.1016/j.urolonc.2019.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
Abstract
This editorial highlights submissions to part II of the 3rd IBCN Seminars Series particularly focusing on the tools required for conduction of translational research in bladder cancer. One of the submissions describe the ex vivo culture of primary tumor cells from N-methyl-N-nitrosourea-induced bladder tumors in rats and subsequent establishment of an immortalized cell line. In a next step the authors thoroughly characterize this cell line. They conclude that differentiation marker expression patterns observed in the original tumors are largely retained in the spheroids. Although new cancer models, such as organoid tissue cultures, hold great promise for studying cancer progression and might have a potential for development and selection of an optimal treatment, their limitations must be kept in mind. The second submission, therefore, critically questions the current role of organoid tissue culture as a predictive tool in urothelial cancer patients. The third manuscript of this series provides a broader overview of post-translational modification in bladder cancer is presented and how PTMs can be exploited as potential therapeutic targets. The 3 manuscripts featured in this issue demonstrate especially how basic research is being channeled to inform clinically actionable discoveries.
Collapse
Affiliation(s)
- Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Peter J Goebell
- Department of Urology and Pediatric Urology, Friedrich-Alexander University, Erlangen, Germany
| | - Ashish M Kamat
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Roland Seiler
- Department of Urology, University of Bern, Bern, Switzerland
| | | | - Bernd J Schmitz-Dräger
- Department of Urology and Pediatric Urology, Friedrich-Alexander University, Erlangen, Germany; MVZ Urologie 24, Nürnberg, Germany.
| |
Collapse
|
17
|
Van Nyen T, Moiola CP, Colas E, Annibali D, Amant F. Modeling Endometrial Cancer: Past, Present, and Future. Int J Mol Sci 2018; 19:E2348. [PMID: 30096949 PMCID: PMC6121384 DOI: 10.3390/ijms19082348] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Endometrial cancer is the most common type of cancer of the female reproductive tract. Although prognosis is generally good for patients with low-grade and early-stage diseases, the outcomes for high-grade and metastatic/recurrent cases remain poor, since traditional chemotherapy regimens based on platinum and taxanes have limited effects. No targeted agents have been approved so far, although several new drugs have been tested without striking results in clinical trials. Over the last decades, many efforts have been made towards the establishment and development of preclinical models, aiming at recapitulating the structural and molecular determinants of the disease. Here, we present an overview of the most commonly used in vitro and in vivo models and discuss their peculiar features, describing their main applications and the value in the advancement of both fundamental and translational endometrial cancer research.
Collapse
Affiliation(s)
- Tom Van Nyen
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
| | - Cristian P Moiola
- Pathological Oncology Group, Biomedical Research Institute of Lleida (IRBLLEIDA), University Hospital Arnau de Vilanova, 25198 Lleida, Spain.
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, CIBERONC, 08035 Barcelona, Spain.
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, CIBERONC, 08035 Barcelona, Spain.
| | - Daniela Annibali
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
| | - Frédéric Amant
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
- Centre for Gynecologic Oncology Amsterdam (CGOA), Antoni Van Leeuwenhoek-Netherlands Cancer Institute (Avl-NKI) and University Medical Centra (UMC), 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Kobayashi PE, Fonseca-Alves CE, Rivera-Calderón LG, Carvalho M, Kuasne H, Rogatto SR, Laufer-Amorim R. Deregulation of E-cadherin, β-catenin, APC and Caveolin-1 expression occurs in canine prostate cancer and metastatic processes. Res Vet Sci 2018; 118:254-261. [PMID: 29529534 DOI: 10.1016/j.rvsc.2018.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/12/2018] [Accepted: 03/02/2018] [Indexed: 01/08/2023]
Abstract
Prostate cancer is a heterogeneous disease with high levels of clinical and gene heterogeneity, consequently offering several targets for therapy. Dogs with naturally occurring prostate cancer are useful models for molecular investigations and studying new treatment efficacy. Three genes and proteins associated with the WNT pathway (β-catenin, APC and E-cadherin) and Caveolin-1 (CAV-1) were evaluated in canine pre-neoplastic proliferative inflammatory atrophy (PIA), prostate cancer and metastatic disease. The APC gene methylation status was also investigated. As in human prostate cancer, cytoplasmic and nuclear β-catenin, which are fundamental for activating the canonical WNT pathway, were found in canine prostate cancer and metastasis. Membranous E-cadherin was also lost in these lesions, allowing cellular migration to the stroma and nuclear localization of β-catenin. In contrast to human prostate tumours, no APC downregulation or hypermethylation was found in canine prostate cancer. The CAV-1 gene and protein overexpression were found in canine prostate cancer, and as in humans, the highest levels were found in Gleason scores ≥8. In conclusion, as with human prostate cancer, β-catenin and E-cadherin in the WNT pathway, as well as Caveolin-1, are molecular drivers in canine prostate cancer. These findings provide additional evidence that dogs are useful models for studying new therapeutic targets in prostate cancer.
Collapse
Affiliation(s)
- Priscila E Kobayashi
- São Paulo State University (UNESP), Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Botucatu, SP, Brazil
| | - Carlos E Fonseca-Alves
- São Paulo State University (UNESP), Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Botucatu, SP, Brazil
| | - Luis G Rivera-Calderón
- São Paulo State University (UNESP), Department of Veterinary Pathology, School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Márcio Carvalho
- São Paulo State University (UNESP), Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Botucatu, SP, Brazil
| | - Hellen Kuasne
- International Center for Research (CIPE), AC Camargo Hospital, Liberdade, São Paulo, Brazil
| | - Silvia R Rogatto
- Department of Clinical Genetics, Vejle Hospital and Institute of Regional Health, University of Southern Denmark, Denmark
| | - Renée Laufer-Amorim
- São Paulo State University (UNESP), Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Botucatu, SP, Brazil.
| |
Collapse
|
19
|
Rubio-Manzanares Dorado M, Marín Gómez LM, Aparicio Sánchez D, Pereira Arenas S, Praena-Fernández JM, Borrero Martín JJ, Farfán López F, Gómez Bravo MÁ, Muntané Relat J, Padillo Ruiz J. Translational pancreatic cancer research: A comparative study on patient-derived xenograft models. World J Gastroenterol 2018; 24:794-809. [PMID: 29467550 PMCID: PMC5807938 DOI: 10.3748/wjg.v24.i7.794] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/14/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the viability of orthotopic and heterotopic patient-derived pancreatic cancer xenografts implanted into nude mice. METHODS This study presents a prospective experimental analytical follow-up of the development of tumours in mice upon implantation of human pancreatic adenocarcinoma samples. Specimens were obtained surgically from patients with a pathological diagnosis of pancreatic adenocarcinoma. Tumour samples from pancreatic cancer patients were transplanted into nude mice in three different locations (intraperitoneal, subcutaneous and pancreatic). Histological analysis (haematoxylin-eosin and Masson's trichrome staining) and immunohistochemical assessment of apoptosis (TUNEL), proliferation (Ki-67), angiogenesis (CD31) and fibrogenesis (α-SMA) were performed. When a tumour xenograft reached the target size, it was re-implanted in a new nude mouse. Three sequential tumour xenograft generations were generated (F1, F2 and F3). RESULTS The overall tumour engraftment rate was 61.1%. The subcutaneous model was most effective in terms of tissue growth (69.9%), followed by intraperitoneal (57.6%) and pancreatic (55%) models. Tumour development was faster in the subcutaneous model (17.7 ± 2.6 wk) compared with the pancreatic (23.1 ± 2.3 wk) and intraperitoneal (25.0 ± 2.7 wk) models (P = 0.064). There was a progressive increase in the tumour engraftment rate over successive generations for all three models (F1 28.1% vs F2 71.4% vs F3 80.9%, P < 0.001). There were no significant differences in tumour xenograft differentiation and cell proliferation between human samples and the three experimental models among the sequential generations of tumour xenografts. However, a progressive decrease in fibrosis, fibrogenesis, tumour vascularisation and apoptosis was observed in the three experimental models compared with the human samples. All three pancreatic patient-derived xenograft models presented similar histological and immunohistochemical characteristics. CONCLUSION In our experience, the faster development and greatest number of viable xenografts could make the subcutaneous model the best option for experimentation in pancreatic cancer.
Collapse
Affiliation(s)
| | - Luis Miguel Marín Gómez
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Daniel Aparicio Sánchez
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Sheila Pereira Arenas
- Oncology Surgery, Cell Therapy, and Organ Transplantation Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, University of Seville, Seville 41013, Spain
| | - Juan Manuel Praena-Fernández
- Statistics, Methodology and Evaluation of Research Unit, Virgen del Rocio University Hospital, Seville 41013, Spain
| | | | | | - Miguel Ángel Gómez Bravo
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Jordi Muntané Relat
- Oncology Surgery, Cell Therapy, and Organ Transplantation Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, University of Seville, Seville 41013, Spain
| | - Javier Padillo Ruiz
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| |
Collapse
|
20
|
Yoshida T, Kates M, Sopko NA, Liu X, Singh AK, Bishai WR, Joice G, McConkey DJ, Bivalacqua TJ. Ex vivo culture of tumor cells from N-methyl-N-nitrosourea-induced bladder cancer in rats: Development of organoids and an immortalized cell line. Urol Oncol 2017; 36:160.e23-160.e32. [PMID: 29288005 DOI: 10.1016/j.urolonc.2017.11.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/24/2017] [Accepted: 11/30/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE We ex vivo cultured primary tumor cells from N-methyl-N-nitrosourea (MNU)-induced bladder tumors in rats and established an immortalized cell line from them. MATERIALS AND METHODS Bladder tumors in rats were induced by instillation of MNU into the murine bladder. Primary tumor cells were prepared by the cancer-tissue originated spheroid method. An immortalized cell line was established by co-culture with fibroblasts. The cultured tumor cells were molecularly and functionally characterized by quantitative real-time polymerase chain reaction, Western blot, growth assay, and transwell migration assay. RESULTS Primary tumor cells were successfully prepared as multicellular spheroids from MNU-induced bladder tumors. The differentiation marker expression patterns observed in the original tumors were largely retained in the spheroids. We succeeded in establishing a cell line from the spheroids and named it T-MNU-1. Although basal markers (CK14 and CK5) were enriched in T-MNU-1 compared to the spheroids, T-MNU-1 expressed both luminal and basal markers. T-MNU-1 was able to migrate through a transwell. CONCLUSIONS Tumor cells in MNU-induced bladder tumors were successfully cultured ex vivo as organoids, and an immortalized cell line was also established from them. The ex vivo models offer a platform that enables analysis of intrinsic characteristics of tumor cells excluding influence of microenvironment in MNU-induced bladder tumors.
Collapse
Affiliation(s)
- Takahiro Yoshida
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Max Kates
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Nikolai A Sopko
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Xiaopu Liu
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Alok K Singh
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD
| | - William R Bishai
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD
| | - Gregory Joice
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - David J McConkey
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD; The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, MD
| | - Trinity J Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD; The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, MD
| |
Collapse
|
21
|
Gengenbacher N, Singhal M, Augustin HG. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat Rev Cancer 2017; 17:751-765. [PMID: 29077691 DOI: 10.1038/nrc.2017.92] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oncology research in humans is limited to analytical and observational studies for obvious ethical reasons, with therapy-focused clinical trials being the one exception to this rule. Preclinical mouse tumour models therefore serve as an indispensable intermediate experimental model system bridging more reductionist in vitro research with human studies. Based on a systematic survey of preclinical mouse tumour studies published in eight scientific journals in 2016, this Analysis provides an overview of how contemporary preclinical mouse tumour biology research is pursued. It thereby identifies some of the most important challenges in this field and discusses potential ways in which preclinical mouse tumour models could be improved for better relevance, reproducibility and translatability.
Collapse
Affiliation(s)
- Nicolas Gengenbacher
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Cancer Consortium, 69120 Heidelberg, Germany
| |
Collapse
|