1201
|
Grassi L, Alfonsi R, Francescangeli F, Signore M, De Angelis ML, Addario A, Costantini M, Flex E, Ciolfi A, Pizzi S, Bruselles A, Pallocca M, Simone G, Haoui M, Falchi M, Milella M, Sentinelli S, Di Matteo P, Stellacci E, Gallucci M, Muto G, Tartaglia M, De Maria R, Bonci D. Organoids as a new model for improving regenerative medicine and cancer personalized therapy in renal diseases. Cell Death Dis 2019; 10:201. [PMID: 30814510 PMCID: PMC6393468 DOI: 10.1038/s41419-019-1453-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/24/2022]
Abstract
The pressure towards innovation and creation of new model systems in regenerative medicine and cancer research has fostered the development of novel potential therapeutic applications. Kidney injuries provoke a high request of organ transplants making it the most demanding system in the field of regenerative medicine. Furthermore, renal cancer frequently threaten patients’ life and aggressive forms still remain difficult to treat. Ethical issues related to the use of embryonic stem cells, has fueled research on adult, patient-specific pluripotent stem cells as a model for discovery and therapeutic development, but to date, normal and cancerous renal experimental models are lacking. Several research groups are focusing on the development of organoid cultures. Since organoids mimic the original tissue architecture in vitro, they represent an excellent model for tissue engineering studies and cancer therapy testing. We established normal and tumor renal cell carcinoma organoids previously maintained in a heterogeneous multi-clone stem cell-like enriching medium. Starting from adult normal kidney specimens, we were able to isolate and propagate organoid 3D-structures composed of both differentiated and undifferentiated cells while expressing nephron specific markers. Furthermore, we were capable to establish organoids derived from cancer tissues although with a success rate inferior to that of their normal counterpart. Cancer cultures displayed epithelial and mesenchymal phenotype while retaining tumor specific markers. Of note, tumor organoids recapitulated neoplastic masses when orthotopically injected into immunocompromised mice. Our data suggest an innovative approach of long-term establishment of normal- and cancer-derived renal organoids obtained from cultures of fleshly dissociated adult tissues. Our results pave the way to organ replacement pioneering strategies as well as to new models for studying drug-induced nephrotoxicity and renal diseases. Along similar lines, deriving organoids from renal cancer patients opens unprecedented opportunities for generation of preclinical models aimed at improving therapeutic treatments.
Collapse
Affiliation(s)
- Ludovica Grassi
- IRCCS, Regina Elena National Cancer Institute, Rome, Italy.,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Department of Internal Medicine and Medical Specialties, "La Sapienza" University, Rome, Italy
| | - Romina Alfonsi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Rome, Italy.,Istituto di Patologia Generale Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | | | - Michele Signore
- RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio Addario
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Manuela Costantini
- Oncological Urology Department, Regina Elena National Cancer Institute, Rome, Italy.,Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Simone Pizzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Giuseppe Simone
- Oncological Urology Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Mustapha Haoui
- IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine, Verona, Italy.,Verona University, Hospital Trust, Verona, Italy
| | | | - Paola Di Matteo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Emilia Stellacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Michele Gallucci
- Oncological Urology Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Muto
- Department of Urology, Humanitas University, Turin, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Ruggero De Maria
- Istituto di Patologia Generale Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy. .,Scientific Vice-Direction, Fondazione Policlinico Universitario "A. Gemelli" - I.R.C.C.S. Largo Francesco Vito 1-8, 00168, Rome, Italy.
| | - Désirée Bonci
- IRCCS, Regina Elena National Cancer Institute, Rome, Italy. .,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
1202
|
Pancreatic Progenitors and Organoids as a Prerequisite to Model Pancreatic Diseases and Cancer. Stem Cells Int 2019; 2019:9301382. [PMID: 30930950 PMCID: PMC6410438 DOI: 10.1155/2019/9301382] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are characterized by their unique capacity to stepwise differentiate towards any particular cell type in an adult organism. Pluripotent stem cells provide a beneficial platform to model hereditary diseases and even cancer development. While the incidence of pancreatic diseases such as diabetes and pancreatitis is increasing, the understanding of the underlying pathogenesis of particular diseases remains limited. Only a few recent publications have contributed to the characterization of human pancreatic development in the fetal stage. Hence, most knowledge of pancreatic specification is based on murine embryology. Optimizing and understanding current in vitro protocols for pancreatic differentiation of ESCs and iPSCs constitutes a prerequisite to generate functional pancreatic cells for better disease modeling and drug discovery. Moreover, human pancreatic organoids derived from pluripotent stem cells, organ-restricted stem cells, and tumor samples provide a powerful technology to model carcinogenesis and hereditary diseases independent of genetically engineered mouse models. Herein, we summarize recent advances in directed differentiation of pancreatic organoids comprising endocrine cell types. Beyond that, we illustrate up-and-coming applications for organoid-based platforms.
Collapse
|
1203
|
Phan N, Hong JJ, Tofig B, Mapua M, Elashoff D, Moatamed NA, Huang J, Memarzadeh S, Damoiseaux R, Soragni A. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun Biol 2019; 2:78. [PMID: 30820473 PMCID: PMC6389967 DOI: 10.1038/s42003-019-0305-x] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/15/2019] [Indexed: 12/25/2022] Open
Abstract
Tumor organoids maintain cell-cell interactions, heterogeneity, microenvironment, and drug response of the sample they originate from. Thus, there is increasing interest in developing tumor organoid models for drug development and personalized medicine applications. Although organoids are in principle amenable to high-throughput screenings, progress has been hampered by technical constraints and extensive manipulations required by current methods. Here we introduce a miniaturized method that uses a simplified geometry by seeding cells around the rim of the wells (mini-rings). This allows high-throughput screenings in a format compatible with automation as shown using four patient-derived tumor organoids established from two ovarian and one peritoneal high-grade serous carcinomas and one carcinosarcoma of the ovary. Using our automated screening platform, we identified personalized responses by measuring viability, number, and size of organoids after exposure to 240 kinase inhibitors. Results are available within a week from surgery, a timeline compatible with therapeutic decision-making.
Collapse
Affiliation(s)
- Nhan Phan
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, HCM City, Vietnam
| | - Jenny J Hong
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Bobby Tofig
- Molecular Screening Shared Resource, California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
| | - Matthew Mapua
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - David Elashoff
- Department of Biostatistics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Neda A Moatamed
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jin Huang
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA
- The VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Molecular Screening Shared Resource, California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Molecular and Medicinal Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alice Soragni
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
1204
|
Xu LL, Hai P, Zhang SB, Xiao JF, Gao Y, Ma BJ, Fu HY, Chen YM, Yang XL. Prenylated Indole Diterpene Alkaloids from a Mine-Soil-Derived Tolypocladium sp. JOURNAL OF NATURAL PRODUCTS 2019; 82:221-231. [PMID: 30702286 DOI: 10.1021/acs.jnatprod.8b00589] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Ten new prenylated indole diterpene alkaloids, tolypocladin A-J (1-10), including four chlorinated metabolites, have been isolated from a culture of a mine-soil-derived fungus, Tolypocladium sp. XL115. The structures and absolute configurations of 1-10 were determined by spectroscopic analysis, ECD calculations, and comparison with known compounds. Compounds 1 and 8 displayed significant antimicrobial activities. In addition, compound 1 also showed weak cytotoxic activity against all tested human cancer cell lines and suppressed the growth and viability of the patient-derived HCC cells T1224.
Collapse
Affiliation(s)
- Lu-Lin Xu
- School of Pharmaceutical Sciences , South-Central University for Nationalities , Wuhan 430074 , People's Republic of China
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences , Chongqing University , Chongqing 401331 , People's Republic of China
| | - Ping Hai
- Department of Chemical Engineering , Yibin University , Yibin 644000 , People's Republic of China
| | - Shuai-Bing Zhang
- School of Pharmaceutical Sciences , South-Central University for Nationalities , Wuhan 430074 , People's Republic of China
| | - Jing-Fang Xiao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology of the Ministry of Education of China , Third Military Medical University , Chongqing 400038 , People's Republic of China
| | - Yuan Gao
- Department of Chemical Engineering , Yibin University , Yibin 644000 , People's Republic of China
| | - Bing-Ji Ma
- Department of Traditional Chinese Medicine , Henan Agricultural University , Wenhua Road 12 , Zhengzhou 450002 , People's Republic of China
| | - Hai-Yan Fu
- School of Pharmaceutical Sciences , South-Central University for Nationalities , Wuhan 430074 , People's Republic of China
| | - Ye-Miao Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology of the Ministry of Education of China , Third Military Medical University , Chongqing 400038 , People's Republic of China
| | - Xiao-Long Yang
- School of Pharmaceutical Sciences , South-Central University for Nationalities , Wuhan 430074 , People's Republic of China
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences , Chongqing University , Chongqing 401331 , People's Republic of China
| |
Collapse
|
1205
|
Cruz-Acuña R, García AJ. Engineered materials to model human intestinal development and cancer using organoids. Exp Cell Res 2019; 377:109-114. [PMID: 30794801 DOI: 10.1016/j.yexcr.2019.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/27/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023]
Abstract
Human organoids provide constructive in vitro models of human development and disease, as these recapitulate important morphogenetic and functional features of the tissue and species of origin. However, organoid culture technologies often involve the use of biologically-derived materials (e.g. Matrigel™) that do not allow dissection of the independent contributions of the biochemical and biophysical matrix properties to organoid development. Additionally, their inherent lot-to-lot variability and, in the case of Matrigel™, tumor-derived nature limits their applicability as platforms for drug and tissue transplantation therapies. Here, we highlight recent studies that overcome these limitations through engineering of novel biomaterial platforms that (1) allow to study the independent contributions of physicochemical matrix properties to organoid development and their potential for translational therapies, and (2) better recreate the tumor microenvironment for high-throughput, pre-clinical drug development. These studies illustrate how innovative biomaterial constructs can contribute to the modeling of human development and disease using organoids, and as platforms for development of organoid-based therapies. Finally, we discuss the current limitations of the organoid field and how they can potentially be addressed using engineered biomaterials.
Collapse
Affiliation(s)
- Ricardo Cruz-Acuña
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States; Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| |
Collapse
|
1206
|
Sachs N, Papaspyropoulos A, Zomer-van Ommen DD, Heo I, Böttinger L, Klay D, Weeber F, Huelsz-Prince G, Iakobachvili N, Amatngalim GD, de Ligt J, van Hoeck A, Proost N, Viveen MC, Lyubimova A, Teeven L, Derakhshan S, Korving J, Begthel H, Dekkers JF, Kumawat K, Ramos E, van Oosterhout MF, Offerhaus GJ, Wiener DJ, Olimpio EP, Dijkstra KK, Smit EF, van der Linden M, Jaksani S, van de Ven M, Jonkers J, Rios AC, Voest EE, van Moorsel CH, van der Ent CK, Cuppen E, van Oudenaarden A, Coenjaerts FE, Meyaard L, Bont LJ, Peters PJ, Tans SJ, van Zon JS, Boj SF, Vries RG, Beekman JM, Clevers H. Long-term expanding human airway organoids for disease modeling. EMBO J 2019; 38:e100300. [PMID: 30643021 PMCID: PMC6376275 DOI: 10.15252/embj.2018100300] [Citation(s) in RCA: 638] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/30/2022] Open
Abstract
Organoids are self-organizing 3D structures grown from stem cells that recapitulate essential aspects of organ structure and function. Here, we describe a method to establish long-term-expanding human airway organoids from broncho-alveolar resections or lavage material. The pseudostratified airway organoids consist of basal cells, functional multi-ciliated cells, mucus-producing secretory cells, and CC10-secreting club cells. Airway organoids derived from cystic fibrosis (CF) patients allow assessment of CFTR function in an organoid swelling assay. Organoids established from lung cancer resections and metastasis biopsies retain tumor histopathology as well as cancer gene mutations and are amenable to drug screening. Respiratory syncytial virus (RSV) infection recapitulates central disease features, dramatically increases organoid cell motility via the non-structural viral NS2 protein, and preferentially recruits neutrophils upon co-culturing. We conclude that human airway organoids represent versatile models for the in vitro study of hereditary, malignant, and infectious pulmonary disease.
Collapse
Affiliation(s)
- Norman Sachs
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | | | | | - Inha Heo
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Lena Böttinger
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Dymph Klay
- St. Antonius Hospital Nieuwegein, Nieuwegein, The Netherlands
| | - Fleur Weeber
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | - Natalie Proost
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Anna Lyubimova
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Luc Teeven
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Sepideh Derakhshan
- Wilhelmina Children's Hospital and UMC Utrecht, Utrecht, The Netherlands
| | - Jeroen Korving
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Harry Begthel
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Johanna F Dekkers
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | - Kuldeep Kumawat
- Wilhelmina Children's Hospital and UMC Utrecht, Utrecht, The Netherlands
| | - Emilio Ramos
- Hubrecht Organoid Technology, Utrecht, The Netherlands
| | | | | | - Dominique J Wiener
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
| | | | | | - Egbert F Smit
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Emile E Voest
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | - Linde Meyaard
- Wilhelmina Children's Hospital and UMC Utrecht, Utrecht, The Netherlands
| | - Louis J Bont
- Wilhelmina Children's Hospital and UMC Utrecht, Utrecht, The Netherlands
| | | | | | | | - Sylvia F Boj
- Hubrecht Organoid Technology, Utrecht, The Netherlands
| | | | - Jeffrey M Beekman
- Wilhelmina Children's Hospital and UMC Utrecht, Utrecht, The Netherlands
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute-KNAW and UMC Utrecht, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
1207
|
Klose J, Trefz S, Wagner T, Steffen L, Preißendörfer Charrier A, Radhakrishnan P, Volz C, Schmidt T, Ulrich A, Dieter SM, Ball C, Glimm H, Schneider M. Salinomycin: Anti-tumor activity in a pre-clinical colorectal cancer model. PLoS One 2019; 14:e0211916. [PMID: 30763370 PMCID: PMC6375586 DOI: 10.1371/journal.pone.0211916] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Objectives Salinomycin is a polyether antibiotic with selective activity against human cancer stem cells. The impact of salinomycin on patient-derived primary human colorectal cancer cells has not been investigated so far. Thus, here we aimed to investigate the activity of salinomycin against tumor initiating cells isolated from patients with colorectal cancer. Methods Primary tumor-initiating cells (TIC) isolated from human patients with colorectal liver metastases or from human primary colon carcinoma were exposed to salinomycin and compared to treatment with 5-FU and oxaliplatin. TICs were injected subcutaneously into NOD/SCID mice to induce a patient-derived mouse xenograft model of colorectal cancer. Animals were treated either with salinomycin, FOLFOX regimen, or salinomycin and FOLFOX. Human colorectal cancer cells were used to delineate an underlying molecular mechanism of salinomycin in this tumor entity. Results Applying TICs isolated from human patients with colorectal liver metastases or from human primary colon carcinoma, we demonstrated that salinomycin exerts increased antiproliferative activity compared to 5-fluorouracil and oxaliplatin treatment. Consistently, salinomycin alone or in combination with FOLFOX exerts superior antitumor activity compared to FOLFOX therapy in a patient-derived mouse xenograft model of colorectal cancer. Salinomycin induces apoptosis of human colorectal cancer cells, accompanied by accumulation of dysfunctional mitochondria and reactive oxygen species. These effects are associated with expressional down-regulation of superoxide dismutase-1 (SOD1) in response to salinomycin treatment. Conclusion Collectively, the results of this pre-clinical study indicate that salinomycin alone or in combination with 5-fluorouracil and oxaliplatin exerts increased antitumoral activity compared to common chemotherapy.
Collapse
Affiliation(s)
- Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| | - Stefan Trefz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Tobias Wagner
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Luca Steffen
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Claudia Volz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sebastian M. Dieter
- Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), Dresden, Germany
| | - Hanno Glimm
- Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), Dresden, Germany
- Center for Personalized Oncology, University Hospital Carl Gustav Carus Dresden at TU Dresden, Dresden, Germany
- German Consortium for Translational Cancer Research (DKTK) Dresden, Dresden, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
1208
|
Affiliation(s)
- Mo Li
- From the King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (M.L.); and the Salk Institute for Biological Studies, La Jolla, CA (J.C.I.B.)
| | - Juan C Izpisua Belmonte
- From the King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (M.L.); and the Salk Institute for Biological Studies, La Jolla, CA (J.C.I.B.)
| |
Collapse
|
1209
|
Abstract
Cancer research relies on model systems, which reflect the biology of actual human tumours to only a certain extent. One important feature of human cancer is its intra-tumour genomic heterogeneity and instability. However, the extent of such genomic instability in cancer models has received limited attention in research. Here, we review the state of knowledge of genomic instability of cancer models and discuss its biological origins and implications for basic research and for cancer precision medicine. We discuss strategies to cope with such genomic evolution and evaluate both the perils and the emerging opportunities associated with it.
Collapse
Affiliation(s)
- Uri Ben-David
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Rameen Beroukhim
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Brigham and Women's Hospital, Boston, MA, USA.
| | - Todd R Golub
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
1210
|
Seidlitz T, Merker SR, Rothe A, Zakrzewski F, von Neubeck C, Grützmann K, Sommer U, Schweitzer C, Schölch S, Uhlemann H, Gaebler AM, Werner K, Krause M, Baretton GB, Welsch T, Koo BK, Aust DE, Klink B, Weitz J, Stange DE. Human gastric cancer modelling using organoids. Gut 2019; 68:207-217. [PMID: 29703791 PMCID: PMC6352409 DOI: 10.1136/gutjnl-2017-314549] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 04/07/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Gastric cancer is the second leading cause of cancer-related deaths and the fifth most common malignancy worldwide. In this study, human and mouse gastric cancer organoids were generated to model the disease and perform drug testing to delineate treatment strategies. DESIGN Human gastric cancer organoid cultures were established, samples classified according to their molecular profile and their response to conventional chemotherapeutics tested. Targeted treatment was performed according to specific druggable mutations. Mouse gastric cancer organoid cultures were generated carrying molecular subtype-specific alterations. RESULTS Twenty human gastric cancer organoid cultures were established and four selected for a comprehensive in-depth analysis. Organoids demonstrated divergent growth characteristics and morphologies. Immunohistochemistry showed similar characteristics to the corresponding primary tissue. A divergent response to 5-fluoruracil, oxaliplatin, irinotecan, epirubicin and docetaxel treatment was observed. Whole genome sequencing revealed a mutational spectrum that corresponded to the previously identified microsatellite instable, genomic stable and chromosomal instable subtypes of gastric cancer. The mutational landscape allowed targeted therapy with trastuzumab for ERBB2 alterations and palbociclib for CDKN2A loss. Mouse cancer organoids carrying Kras and Tp53 or Apc and Cdh1 mutations were characterised and serve as model system to study the signalling of induced pathways. CONCLUSION We generated human and mouse gastric cancer organoids modelling typical characteristics and altered pathways of human gastric cancer. Successful interference with activated pathways demonstrates their potential usefulness as living biomarkers for therapy response testing.
Collapse
Affiliation(s)
- Therese Seidlitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Sebastian R Merker
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Alexander Rothe
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Falk Zakrzewski
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany
| | - Cläre von Neubeck
- Department of Radiotherapy and Radiation Oncology and National Center for Radiation Research in Oncology (OncoRay), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Konrad Grützmann
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany
| | - Ulrich Sommer
- Institute for Pathology and Tumour and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Christine Schweitzer
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schölch
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Heike Uhlemann
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Anne-Marlene Gaebler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Kristin Werner
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology and National Center for Radiation Research in Oncology (OncoRay), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Gustavo B Baretton
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany,Institute for Pathology and Tumour and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Daniela E Aust
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany,Institute for Pathology and Tumour and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany,Institute for Clinical Genetics, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Daniel E Stange
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany,Partner Site Dresden, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
1211
|
Sun W, Luo Z, Lee J, Kim HJ, Lee K, Tebon P, Feng Y, Dokmeci MR, Sengupta S, Khademhosseini A. Organ-on-a-Chip for Cancer and Immune Organs Modeling. Adv Healthc Mater 2019; 8:e1801363. [PMID: 30605261 PMCID: PMC6424124 DOI: 10.1002/adhm.201801363] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/07/2018] [Indexed: 12/21/2022]
Abstract
Bridging the gap between findings in preclinical 2D cell culture models and in vivo tissue cultures has been challenging; the simple microenvironment of 2D monolayer culture systems may not capture the cellular response to drugs accurately. Three-dimensional organotypic models have gained increasing interest due to their ability to recreate precise cellular organizations. These models facilitate investigation of the interactions between different sub-tissue level components through providing physiologically relevant microenvironments for cells in vitro. The incorporation of human-sourced tissues into these models further enables personalized prediction of drug responses. Integration of microfluidic units into the 3D models can be used to control their local environment, dynamic simulation of cell behaviors, and real-time readout of drug testing data. Cancer and immune system related diseases are severe burdens to our health care system and have created an urgent need for high-throughput, and effective drug development plans. This review focuses on recent progress in the development of "cancer-on-a-chip" and "immune organs-on-a-chip" systems designed to study disease progression and predict drug-induced responses. Future challenges and opportunities are also discussed.
Collapse
Affiliation(s)
- Wujin Sun
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Zhimin Luo
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Junmin Lee
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Han-Jun Kim
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - KangJu Lee
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Peyton Tebon
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Yudi Feng
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; College of Chemistry, Nankai University, Tianjin 300071, China
| | - Mehmet R. Dokmeci
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Shiladitya Sengupta
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, ; Harvard – MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California - Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90024, USA.; Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA; Department of Radiology, University of California-Los Angeles, Los Angeles, CA 90095, USA; Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
1212
|
Fujii M, Clevers H, Sato T. Modeling Human Digestive Diseases With CRISPR-Cas9-Modified Organoids. Gastroenterology 2019; 156:562-576. [PMID: 30476497 DOI: 10.1053/j.gastro.2018.11.048] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/10/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
Abstract
Insights into the stem cell niche have allowed researchers to cultivate adult tissue stem cells as organoids that display structural and phenotypic features of healthy and diseased epithelial tissues. Organoids derived from patients' tissues are used as models of disease and to test drugs. CRISPR-Cas9 technology can be used to genetically engineer organoids for studies of monogenic diseases and cancer. We review the derivation of organoids from human gastrointestinal tissues and how CRISPR-Cas9 technology can be used to study these organoids. We discuss burgeoning technologies that are broadening our understanding of diseases of the digestive system.
Collapse
Affiliation(s)
- Masayuki Fujii
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Hans Clevers
- Hubrecht Institute, University Medical Center Utrecht and Princess Maxima Center, Utrecht, The Netherlands
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
1213
|
Intestinal organoids: A new paradigm for engineering intestinal epithelium in vitro. Biomaterials 2019; 194:195-214. [DOI: 10.1016/j.biomaterials.2018.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/22/2018] [Accepted: 12/08/2018] [Indexed: 12/11/2022]
|
1214
|
Tseng YY, Boehm JS. From cell lines to living biosensors: new opportunities to prioritize cancer dependencies using ex vivo tumor cultures. Curr Opin Genet Dev 2019; 54:33-40. [DOI: 10.1016/j.gde.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 02/23/2019] [Indexed: 01/05/2023]
|
1215
|
Dama E, Melocchi V, Colangelo T, Cuttano R, Bianchi F. Deciphering the Molecular Profile of Lung Cancer: New Strategies for the Early Detection and Prognostic Stratification. J Clin Med 2019; 8:jcm8010108. [PMID: 30658453 PMCID: PMC6352200 DOI: 10.3390/jcm8010108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Recent advances in radiological imaging and genomic analysis are profoundly changing the way to manage lung cancer patients. Screening programs which couple lung cancer risk prediction models and low-dose computed tomography (LDCT) recently showed their effectiveness in the early diagnosis of lung tumors. In addition, the emerging field of radiomics is revolutionizing the approach to handle medical images, i.e., from a “simple” visual inspection to a high-throughput analysis of hundreds of quantitative features of images which can predict prognosis and therapy response. Yet, with the advent of next-generation sequencing (NGS) and the establishment of large genomic consortia, the whole mutational and transcriptomic profile of lung cancer has been unveiled and made publicly available via web services interfaces. This has tremendously accelerated the discovery of actionable mutations, as well as the identification of cancer biomarkers, which are pivotal for development of personalized targeted therapies. In this review, we will describe recent advances in cancer biomarkers discovery for early diagnosis, prognosis, and prediction of chemotherapy response.
Collapse
Affiliation(s)
- Elisa Dama
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Oncology Biomarkers, 71013 San Giovanni Rotondo (FG), Italy.
| | - Valentina Melocchi
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Oncology Biomarkers, 71013 San Giovanni Rotondo (FG), Italy.
| | - Tommaso Colangelo
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Oncology Biomarkers, 71013 San Giovanni Rotondo (FG), Italy.
| | - Roberto Cuttano
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Oncology Biomarkers, 71013 San Giovanni Rotondo (FG), Italy.
| | - Fabrizio Bianchi
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Oncology Biomarkers, 71013 San Giovanni Rotondo (FG), Italy.
| |
Collapse
|
1216
|
Ashley N, Ouaret D, Bodmer WF. Cellular polarity modulates drug resistance in primary colorectal cancers via orientation of the multidrug resistance protein ABCB1. J Pathol 2019; 247:293-304. [PMID: 30306567 PMCID: PMC6519031 DOI: 10.1002/path.5179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/30/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022]
Abstract
Colonic epithelial cells are highly polarised with a lumen‐facing apical membrane, termed the brush border, and a basal membrane in contact with the underlying extracellular matrix (ECM). This polarity is often maintained in cancer tissue in the form of neoplastic glands and has prognostic value. We compared the cellular polarity of several ex vivo spheroid colonic cancer cultures with their parental tumours and found that those grown as non‐attached colonies exhibited apical brush border proteins on their outer cellular membranes. Transfer of these cultures to an ECM, such as collagen, re‐established the centralised apical polarity observed in vivo. The multidrug resistance protein ABCB1 also became aberrantly polarised to outer colony membranes in suspension cultures, unlike cultures grown in collagen, where it was polarised to central lumens. This polarity switch was dependent on the presence of serum or selected serum components, including epidermal growth factor (EGF), transforming growth factor‐β1 (TGF‐β1) and insulin‐like growth factor‐1 (IGF‐1). The apical/basal orientation of primary cancer colon cultures cultured in collagen/serum was modulated by α2β1 integrin signalling. The polarisation of ABCB1 in colonies significantly altered drug uptake and sensitivity, as the outward polarisation of ABCB1 in suspension colonies effluxed substrates more effectively than ECM‐grown colonies with ABCB1 polarised to central lumens. Thus, serum‐free suspension colonies were more resistant to a variety of anti‐cancer drugs than ECM‐grown colonies. In conclusion, the local stroma, or absence thereof, can have profound effects on the sensitivity of colorectal cultures to drugs that are ABCB1 substrates. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Neil Ashley
- Cancer and Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.,Single Cell Genomics Facility, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Djamila Ouaret
- Cancer and Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Walter F Bodmer
- Cancer and Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
1217
|
Sun W, Lee J, Zhang S, Benyshek C, Dokmeci MR, Khademhosseini A. Engineering Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801039. [PMID: 30643715 PMCID: PMC6325626 DOI: 10.1002/advs.201801039] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/10/2018] [Indexed: 05/18/2023]
Abstract
Advances in genomic sequencing and bioinformatics have led to the prospect of precision medicine where therapeutics can be advised by the genetic background of individuals. For example, mapping cancer genomics has revealed numerous genes that affect the therapeutic outcome of a drug. Through materials and cell engineering, many opportunities exist for engineers to contribute to precision medicine, such as engineering biosensors for diagnosis and health status monitoring, developing smart formulations for the controlled release of drugs, programming immune cells for targeted cancer therapy, differentiating pluripotent stem cells into desired lineages, fabricating bioscaffolds that support cell growth, or constructing "organs-on-chips" that can screen the effects of drugs. Collective engineering efforts will help transform precision medicine into a more personalized and effective healthcare approach. As continuous progress is made in engineering techniques, more tools will be available to fully realize precision medicine's potential.
Collapse
Affiliation(s)
- Wujin Sun
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Junmin Lee
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Shiming Zhang
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Cole Benyshek
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Mehmet R. Dokmeci
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California–Los Angeles10833 Le Conte AveLos AngelesCA90024USA
- Department of Chemical and Biomolecular EngineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center of NanotechnologyDepartment of PhysicsKing Abdulaziz UniversityJeddah21569Saudi Arabia
- Department of Bioindustrial TechnologiesCollege of Animal Bioscience and TechnologyKonkuk UniversitySeoul05029Republic of Korea
| |
Collapse
|
1218
|
Abstract
The adult gastrointestinal tract (GI) is a series of connected organs (esophagus, stomach, small intestine, colon) that develop via progressive regional specification of a continuous tubular embryonic organ anlage. This chapter focuses on organogenesis of the small intestine. The intestine arises by folding of a flat sheet of endodermal cells into a tube of highly proliferative pseudostratified cells. Dramatic elongation of this tube is driven by rapid epithelial proliferation. Then, epithelial-mesenchymal crosstalk and physical forces drive a stepwise cascade that results in convolution of the tubular surface into finger-like projections called villi. Concomitant with villus formation, a sharp epithelial transcriptional boundary is defined between stomach and intestine. Finally, flask-like depressions called crypts are established to house the intestinal stem cells needed throughout life for epithelial renewal. New insights into these events are being provided by in vitro organoid systems, which hold promise for future regenerative engineering of the small intestine.
Collapse
Affiliation(s)
- Sha Wang
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| | - Katherine D Walton
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States.
| | - Deborah L Gumucio
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| |
Collapse
|
1219
|
Abstract
The intestinal epithelium withstands continuous mechanical, chemical and biological insults despite its single-layered, simple epithelial structure. The crypt-villus tissue architecture in combination with rapid cell turnover enables the intestine to act both as a barrier and as the primary site of nutrient uptake. Constant tissue replenishment is fuelled by continuously dividing stem cells that reside at the bottom of crypts. These cells are nurtured and protected by specialized epithelial and mesenchymal cells, and together constitute the intestinal stem cell niche. Intestinal stem cells and early progenitor cells compete for limited niche space and, therefore, the ability to retain or regain stemness. Those cells unable to do so differentiate to one of six different mature cell types and move upwards towards the villus, where they are shed into the intestinal lumen after 3-5 days. In this Review, we discuss the signals, cell types and mechanisms that control homeostasis and regeneration in the intestinal epithelium. We investigate how the niche protects and instructs intestinal stem cells, which processes drive differentiation of mature cells and how imbalance in key signalling pathways can cause human disease.
Collapse
|
1220
|
Chen HI, Song H, Ming GL. Applications of Human Brain Organoids to Clinical Problems. Dev Dyn 2019; 248:53-64. [PMID: 30091290 PMCID: PMC6312736 DOI: 10.1002/dvdy.24662] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are an exciting new technology with the potential to significantly change how diseases of the brain are understood and treated. These three-dimensional neural tissues are derived from the self-organization of pluripotent stem cells, and they recapitulate the developmental process of the human brain, including progenitor zones and rudimentary cortical layers. Brain organoids have been valuable in investigating different aspects of developmental neurobiology and comparative biology. Several characteristics of organoids also make them attractive as models of brain disorders. Data generated from human organoids are more generalizable to patients because of the match in species background. Personalized organoids also can be generated from patient-derived induced pluripotent stem cells. Furthermore, the three-dimensionality of brain organoids supports cellular, mechanical, and topographical cues that are lacking in planar systems. In this review, we discuss the translational potential of brain organoids, using the examples of Zika virus, autism-spectrum disorder, and glioblastoma multiforme to consider how they could contribute to disease modeling, personalized medicine, and testing of therapeutics. We then discuss areas of improvement in organoid technology that will enhance the translational potential of brain organoids, as well as the possibility of their use as substrates for repairing cerebral circuitry after injury. Developmental Dynamics 248:53-64, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- H. Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Guo-li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
1221
|
Murray G, Turner TH, Leslie KA, Alzubi MA, Guest D, Sohal SS, Teitell MA, Harrell JC, Reed J. Live Cell Mass Accumulation Measurement Non-Invasively Predicts Carboplatin Sensitivity in Triple-Negative Breast Cancer Patient-Derived Xenografts. ACS OMEGA 2018; 3:17687-17692. [PMID: 30613814 PMCID: PMC6312628 DOI: 10.1021/acsomega.8b02224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/29/2018] [Indexed: 05/30/2023]
Abstract
Prompt and repeated assessments of tumor sensitivity to available therapeutics could reduce patient morbidity and mortality by quickly identifying therapeutic resistance and optimizing treatment regimens. Analysis of changes in cancer cell biomass has shown promise in assessing drug sensitivity and fulfilling these requirements. However, a major limitation of previous studies in solid tumors, which comprise 90% of cancers, is the use of cancer cell lines rather than freshly isolated tumor material. As a result, existing biomass protocols are not obviously extensible to real patient tumors owing to potential artifacts that would be generated by the removal of cells from their microenvironment and the deleterious effects of excision and purification. In this present work, we show that simple excision of human triple-negative breast cancer (TNBC) tumors growing in immunodeficient mouse, patient-derived xenograft (PDX) models, followed by enzymatic disaggregation into single cell suspension, is enabling for rapid and accurate biomass accumulation-based predictions of in vivo sensitivity to the chemotherapeutic drug carboplatin. We successfully correlate in vitro biomass results with in vivo treatment results in three TNBC PDX models that have differential sensitivity to this drug. With a maximum turnaround time of 40 h from tumor excision to useable results and a fully-automated analysis pipeline, the assay described here has significant potential for translation to clinical practice.
Collapse
Affiliation(s)
- Graeme
F. Murray
- Department
of Physics, Virginia Commonwealth University, 701 West Grace Street, Richmond, Virginia 23284, United States
| | - Tia H. Turner
- Department
of Pathology, Virginia Commonwealth University, 401 North 13th Street, Richmond, Virginia 23298, United States
- Wright
Center for Clinical and Translational Research, Virginia Commonwealth University, 1200 East Clay Street, Richmond, Virginia 23298, United States
| | - Kevin A. Leslie
- Department
of Physics, Virginia Commonwealth University, 701 West Grace Street, Richmond, Virginia 23284, United States
| | - Mohammad A. Alzubi
- Department
of Pathology, Virginia Commonwealth University, 401 North 13th Street, Richmond, Virginia 23298, United States
| | - Daniel Guest
- Department
of Physics, Virginia Commonwealth University, 701 West Grace Street, Richmond, Virginia 23284, United States
| | - Sahib S. Sohal
- Department
of Pathology, Virginia Commonwealth University, 401 North 13th Street, Richmond, Virginia 23298, United States
| | - Michael A. Teitell
- Department
of Pathology and Laboratory Medicine, University
of California Los Angeles, 757 Westwood Plaza, Los Angeles, California 90095, United States
| | - J. Chuck Harrell
- Department
of Pathology, Virginia Commonwealth University, 401 North 13th Street, Richmond, Virginia 23298, United States
- Wright
Center for Clinical and Translational Research, Virginia Commonwealth University, 1200 East Clay Street, Richmond, Virginia 23298, United States
- Massey
Cancer Center, Virginia Commonwealth University, 401 College Street, Box 980037, Richmond, Virginia 23298, United States
| | - Jason Reed
- Department
of Physics, Virginia Commonwealth University, 701 West Grace Street, Richmond, Virginia 23284, United States
- Massey
Cancer Center, Virginia Commonwealth University, 401 College Street, Box 980037, Richmond, Virginia 23298, United States
| |
Collapse
|
1222
|
Cho SJ, Yoon C, Lee JH, Chang KK, Lin JX, Kim YH, Kook MC, Aksoy BA, Park DJ, Ashktorab H, Smoot DT, Schultz N, Yoon SS. RETRACTED: KMT2C Mutations in Diffuse-Type Gastric Adenocarcinoma Promote Epithelial-to-Mesenchymal Transition. Clin Cancer Res 2018; 24:6556-6569. [PMID: 30108106 PMCID: PMC6295255 DOI: 10.1158/1078-0432.ccr-17-1679] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/13/2017] [Accepted: 08/09/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Lauren diffuse-type gastric adenocarcinomas (DGAs) are generally genomically stable. We identified lysine (K)-specific methyltransferase 2C (KMT2C) as a frequently mutated gene and examined its role in DGA progression. EXPERIMENTAL DESIGN We performed whole exome sequencing on tumor samples of 27 patients with DGA who underwent gastrectomy. Lysine (K)-specific methyltransferase 2C (KMT2C) was analyzed in DGA cell lines and in patient tumors. RESULTS KMT2C was the most frequently mutated gene (11 of 27 tumors [41%]). KMT2C expression by immunohistochemistry in tumors from 135 patients with DGA undergoing gastrectomy inversely correlated with more advanced tumor stage (P = 0.023) and worse overall survival (P = 0.017). KMT2C shRNA knockdown in non-transformed HFE-145 gastric epithelial cells promoted epithelial-to-mesenchymal transition (EMT) as demonstrated by increased expression of EMT-related proteins N-cadherin and Slug. Migration and invasion in gastric epithelial cells following KMT2C knockdown increased by 47- to 88-fold. In the DGA cell lines MKN-45 and SNU-668, which have lost KMT2C expression, KMT2C re-expression decreased expression of EMT-related proteins, reduced cell migration by 52% to 60%, and reduced cell invasion by 50% to 74%. Flank xenografts derived from KMT2C-expressing DGA organoids, compared with wild-type organoids, grew more slowly and lost their infiltrative leading edge. EMT can lead to the acquisition of cancer stem cell (CSC) phenotypes. KMT2C re-expression in DGA cell lines reduced spheroid formation by 77% to 78% and reversed CSC resistance to chemotherapy via promotion of DNA damage and apoptosis. CONCLUSIONS KMT2C is frequently mutated in certain populations with DGA. KMT2C loss in DGA promotes EMT and is associated with worse overall survival.
Collapse
Affiliation(s)
- Soo-Jeong Cho
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
- Center for Gastric Cancer, National Cancer Center, Goyang, South Korea
| | - Changhwan Yoon
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jun Ho Lee
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kevin K Chang
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jian-Xian Lin
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Young-Ho Kim
- Division of Clinical Research, Rare Cancer Branch, National Cancer Center, Goyang, South Korea
| | - Myeong-Cherl Kook
- Center for Gastric Cancer, National Cancer Center, Goyang, South Korea
| | - Bülent Arman Aksoy
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Do Joong Park
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | - Duane T Smoot
- Department of Internal Medicine, Meharry Medical College, Nashville, Tennessee
| | - Nikolaus Schultz
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Sam S Yoon
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
1223
|
Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, Deutsch BC, Liao L, Zemek AJ, Zhao F, Karlsson K, Schultz LM, Metzner TJ, Nadauld LD, Tseng YY, Alkhairy S, Oh C, Keskula P, Mendoza-Villanueva D, De La Vega FM, Kunz PL, Liao JC, Leppert JT, Sunwoo JB, Sabatti C, Boehm JS, Hahn WC, Zheng GXY, Davis MM, Kuo CJ. Organoid Modeling of the Tumor Immune Microenvironment. Cell 2018; 175:1972-1988.e16. [PMID: 30550791 PMCID: PMC6656687 DOI: 10.1016/j.cell.2018.11.021] [Citation(s) in RCA: 951] [Impact Index Per Article: 135.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/25/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
Abstract
In vitro cancer cultures, including three-dimensional organoids, typically contain exclusively neoplastic epithelium but require artificial reconstitution to recapitulate the tumor microenvironment (TME). The co-culture of primary tumor epithelia with endogenous, syngeneic tumor-infiltrating lymphocytes (TILs) as a cohesive unit has been particularly elusive. Here, an air-liquid interface (ALI) method propagated patient-derived organoids (PDOs) from >100 human biopsies or mouse tumors in syngeneic immunocompetent hosts as tumor epithelia with native embedded immune cells (T, B, NK, macrophages). Robust droplet-based, single-cell simultaneous determination of gene expression and immune repertoire indicated that PDO TILs accurately preserved the original tumor T cell receptor (TCR) spectrum. Crucially, human and murine PDOs successfully modeled immune checkpoint blockade (ICB) with anti-PD-1- and/or anti-PD-L1 expanding and activating tumor antigen-specific TILs and eliciting tumor cytotoxicity. Organoid-based propagation of primary tumor epithelium en bloc with endogenous immune stroma should enable immuno-oncology investigations within the TME and facilitate personalized immunotherapy testing.
Collapse
Affiliation(s)
- James T Neal
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xingnan Li
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Junjie Zhu
- Department of Electrical Engineering, Stanford University School of Engineering, Stanford, CA, USA
| | | | - Caitlin L Grzeskowiak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jihang Ju
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris H Liu
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shin-Heng Chiou
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Ameen A Salahudeen
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amber R Smith
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brian C Deutsch
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lillian Liao
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Allison J Zemek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fan Zhao
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasper Karlsson
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Liora M Schultz
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Metzner
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lincoln D Nadauld
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuen-Yi Tseng
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Coyin Oh
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Paula Keskula
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | | | - Pamela L Kunz
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Liao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - John T Leppert
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - John B Sunwoo
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA; Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA
| | - Jesse S Boehm
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute and Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
1224
|
Sato Y, Bolzenius JK, Eteleeb AM, Su X, Maher CA, Sehn JK, Arora VK. CD4+ T cells induce rejection of urothelial tumors after immune checkpoint blockade. JCI Insight 2018; 3:121062. [PMID: 30518683 PMCID: PMC6328023 DOI: 10.1172/jci.insight.121062] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/31/2018] [Indexed: 12/30/2022] Open
Abstract
Immune checkpoint blockade (ICB) provides clinical benefit to a minority of patients with urothelial carcinoma (UC). The role of CD4+ T cells in ICB-induced antitumor activity is not well defined; however, CD4+ T cells are speculated to play a supportive role in the development of CD8+ T cells that kill tumor cells after recognition of tumor antigens presented by MHC class I. To investigate the mechanisms of ICB-induced activity against UC, we developed mouse organoid-based transplantable models that have histologic and genetic similarity to human bladder cancer. We found that ICB can induce tumor rejection and protective immunity with these systems in a manner dependent on CD4+ T cells but not reliant on CD8+ T cells. Evaluation of tumor infiltrates and draining lymph nodes after ICB revealed expansion of IFN-γ-producing CD4+ T cells. Tumor cells in this system express MHC class I, MHC class II, and the IFN-γ receptor (Ifngr1), but none were necessary for ICB-induced tumor rejection. IFN-γ neutralization blocked ICB activity, and, in mice depleted of CD4+ T cells, IFN-γ ectopically expressed in the tumor microenvironment was sufficient to inhibit growth of tumors in which the epithelial compartment lacked Ifngr1. Our findings suggest unappreciated CD4+ T cell-dependent mechanisms of ICB activity, principally mediated through IFN-γ effects on the microenvironment.
Collapse
Affiliation(s)
- Yuji Sato
- Department of Internal Medicine, Division of Oncology
| | | | | | - Xinming Su
- Department of Internal Medicine, Division of Oncology
| | - Christopher A. Maher
- Department of Internal Medicine, Division of Oncology
- McDonnell Genome Institute, and
| | - Jennifer K. Sehn
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
1225
|
Xu H, Jiao Y, Qin S, Zhao W, Chu Q, Wu K. Organoid technology in disease modelling, drug development, personalized treatment and regeneration medicine. Exp Hematol Oncol 2018; 7:30. [PMID: 30534474 PMCID: PMC6282260 DOI: 10.1186/s40164-018-0122-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022] Open
Abstract
Organoid technology bridges the gap between conventional two-dimensional cell line culture and in vivo models. The near-physiological technology can virtually recapitulates organ development and human diseases, such as infectious diseases, genetic abnormality and even cancers. In addition, organoids can more accurately predict drug responses, and serve as an excellent platform for drug development, including efficacy evaluation, toxicity testing and pharmacokinetics analysis. Furthermore, organoids can also be exploited to explore the possible optimized treatment strategies for each individual patient. Besides, organoid technology is a promising strategy for regeneration medicine and transplantation use, which can overcome the deficiency in the supply of healthy donor tissues and inherent immunological rejection through establishing isogenic organoids from minuscule amounts of patient biopsies. Collectively, organoids hold enormous potential for clinical applications and bring basic research closer to clinical practice. In this review, we described common organoid lines, summarized the potential clinical applications, and outlined the current limitations.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Ying Jiao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
1226
|
Nagle PW, Plukker JTM, Muijs CT, van Luijk P, Coppes RP. Patient-derived tumor organoids for prediction of cancer treatment response. Semin Cancer Biol 2018; 53:258-264. [DOI: 10.1016/j.semcancer.2018.06.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
|
1227
|
Oksdath M, Perrin SL, Bardy C, Hilder EF, DeForest CA, Arrua RD, Gomez GA. Review: Synthetic scaffolds to control the biochemical, mechanical, and geometrical environment of stem cell-derived brain organoids. APL Bioeng 2018; 2:041501. [PMID: 31069322 PMCID: PMC6481728 DOI: 10.1063/1.5045124] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/31/2018] [Indexed: 01/16/2023] Open
Abstract
Stem cell-derived brain organoids provide a powerful platform for systematic studies of tissue functional architecture and the development of personalized therapies. Here, we review key advances at the interface of soft matter and stem cell biology on synthetic alternatives to extracellular matrices. We emphasize recent biomaterial-based strategies that have been proven advantageous towards optimizing organoid growth and controlling the geometrical, biomechanical, and biochemical properties of the organoid's three-dimensional environment. We highlight systems that have the potential to increase the translational value of region-specific brain organoid models suitable for different types of manipulations and high-throughput applications.
Collapse
Affiliation(s)
- Mariana Oksdath
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | - Sally L. Perrin
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | | | - Emily F. Hilder
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Cole A. DeForest
- Department of Chemical Engineering and Department of Bioengineering, University of Washington, Seattle, Washington 98195-1750, USA
| | - R. Dario Arrua
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Guillermo A. Gomez
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
1228
|
Lordick F. Towards risk-adapted perioperative treatment of gastroesophageal cancer. Ann Oncol 2018; 29:2282-2284. [DOI: 10.1093/annonc/mdy416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
1229
|
Mills RJ, Parker BL, Monnot P, Needham EJ, Vivien CJ, Ferguson C, Parton RG, James DE, Porrello ER, Hudson JE. Development of a human skeletal micro muscle platform with pacing capabilities. Biomaterials 2018; 198:217-227. [PMID: 30527761 DOI: 10.1016/j.biomaterials.2018.11.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/28/2018] [Accepted: 11/22/2018] [Indexed: 12/15/2022]
Abstract
Three dimensional engineered culture systems are powerful tools to rapidly expand our knowledge of human biology and identify novel therapeutic targets for disease. Bioengineered skeletal muscle has been recently shown to recapitulate many features of native muscle biology. However, current skeletal muscle bioengineering approaches require large numbers of cells, reagents and labour, limiting their potential for high-throughput studies. Herein, we use a miniaturized 96-well micro-muscle platform to facilitate semi-automated tissue formation, culture and analysis of human skeletal micro muscles (hμMs). Utilising an iterative screening approach we define a serum-free differentiation protocol that drives rapid, directed differentiation of human myoblast to skeletal myofibres. The resulting hμMs comprised organised bundles of striated and functional myofibres, which respond appropriately to electrical stimulation. Additionally, we developed an optogenetic approach to chronically stimulate hμM to recapitulate known features of exercise training including myofibre hypertrophy and increased expression of metabolic proteins. Taken together, our miniaturized approach provides a new platform to enable high-throughput studies of human skeletal muscle biology and exercise physiology.
Collapse
Affiliation(s)
- Richard J Mills
- School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Queensland, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, St Lucia, 4072, Queensland, Australia; QIMR Berghofer Medical Research Institute, Brisbane, 4006, Queensland, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Pauline Monnot
- School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Queensland, Australia; Laboratoire de Biologie du Développement-Institut de Biologie, CNRS, Sorbonne Université, 75005, Paris, France
| | - Elise J Needham
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Celine J Vivien
- School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Queensland, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, St Lucia, 4072, Queensland, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, 4072, Queensland, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, 4072, Queensland, Australia; Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, 4072, Queensland, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Enzo R Porrello
- School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Queensland, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, St Lucia, 4072, Queensland, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia; Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - James E Hudson
- School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Queensland, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, St Lucia, 4072, Queensland, Australia; QIMR Berghofer Medical Research Institute, Brisbane, 4006, Queensland, Australia.
| |
Collapse
|
1230
|
Kondo J, Ekawa T, Endo H, Yamazaki K, Tanaka N, Kukita Y, Okuyama H, Okami J, Imamura F, Ohue M, Kato K, Nomura T, Kohara A, Mori S, Dan S, Inoue M. High-throughput screening in colorectal cancer tissue-originated spheroids. Cancer Sci 2018; 110:345-355. [PMID: 30343529 PMCID: PMC6317944 DOI: 10.1111/cas.13843] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Patient‐derived cancer organoid culture is an important live material that reflects clinical heterogeneity. However, the limited amount of organoids available for each case as well as the considerable amount of time and cost to expand in vitro makes it impractical to perform high‐throughput drug screening using organoid cultures from multiple patients. Here, we report an advanced system for the high‐throughput screening of 2427 drugs using the cancer tissue‐originated spheroid (CTOS) method. In this system, we apply the CTOS method in an ex vivo platform from xenograft tumors, using machines to handle CTOS and reagents, and testing a CTOS reference panel of multiple CTOS lines for the hit drugs. CTOS passages in xenograft tumors resulted in minimal changes of morphological and genomic status, and xenograft tumor generation efficiently expanded the number of CTOS to evaluate multiple drugs. Our panel of colorectal cancer CTOS lines exhibited diverse sensitivities to the hit compounds, demonstrating the usefulness of this system for investigating highly heterogeneous disease.
Collapse
Affiliation(s)
- Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoya Ekawa
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Kanami Yamazaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Norio Tanaka
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Tokyo, Japan
| | - Yoji Kukita
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroaki Okuyama
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Jiro Okami
- Department of Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Fumio Imamura
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masayuki Ohue
- Department of Gastrointestinal Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kikuya Kato
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Taisei Nomura
- Animal Models of Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Arihiro Kohara
- Laboratory of Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
1231
|
Mittal R, Woo FW, Castro CS, Cohen MA, Karanxha J, Mittal J, Chhibber T, Jhaveri VM. Organ‐on‐chip models: Implications in drug discovery and clinical applications. J Cell Physiol 2018; 234:8352-8380. [DOI: 10.1002/jcp.27729] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Frank W. Woo
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Carlo S. Castro
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Madeline A. Cohen
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Joana Karanxha
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Tanya Chhibber
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University Chandigarh India
| | - Vasanti M. Jhaveri
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
1232
|
Perkhofer L, Frappart PO, Müller M, Kleger A. Importance of organoids for personalized medicine. Per Med 2018; 15:461-465. [PMID: 30418092 DOI: 10.2217/pme-2018-0071] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The establishment of organoid culture systems represents a milestone on the route toward successful personalized medicine. This mini review provides an update on the current status of organoid technology and summarizes their applications in personalized medicine. Organoids can be defined as 3D structures derived either from pluripotent or organ restricted stem cells harboring the ability to mimic in vivo architecture and multi lineage differentiation of terminally differentiated tissues. Due to their unique ability of virtually unlimited self-renewal, organoid cultures should be distinguished from previous 'sphere'-culture assays, for example, 'tumor spheres' that have already been described and applied over the last decades.
Collapse
|
1233
|
|
1234
|
Votanopoulos KI, Mazzocchi A, Sivakumar H, Forsythe S, Aleman J, Levine EA, Skardal A. Appendiceal Cancer Patient-Specific Tumor Organoid Model for Predicting Chemotherapy Efficacy Prior to Initiation of Treatment: A Feasibility Study. Ann Surg Oncol 2018; 26:139-147. [PMID: 30414038 DOI: 10.1245/s10434-018-7008-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION We have hypothesized that biofabrication of appendiceal tumor organoids allows for a more personalized clinical approach and facilitates research in a rare disease. METHODS Appendiceal cancer specimens obtained during cytoreduction with hyperthermic intraperitoneal chemotherapy procedures (CRS/HIPEC) were dissociated and incorporated into an extracellular matrix-based hydrogel system as three-dimensional (3D), patient-specific tumor organoids. Cells were not sorted, preserving tumor heterogeneity, including stroma and immune cell components. Following establishment of organoid sets, chemotherapy drugs were screened in parallel. Live/dead staining and quantitative metabolism assays recorded which chemotherapies were most effective in killing cancer cells for a specific patient. Maintenance of cancer phenotypes were confirmed by using immunohistochemistry. RESULTS Biospecimens from 12 patients were applied for organoid development between November 2016 and May 2018. Successful establishment rate of viable organoid sets was 75% (9/12). Average time from organoid development to chemotherapy testing was 7 days. These tumors included three high-grade appendiceal (HGA) and nine low-grade appendiceal (LGA) primaries obtained from sites of peritoneal metastasis. All tumor organoids were tested with chemotherapeutic agents exhibited responses that were either similar to the patient response or within the variability of the expected clinical response. More specifically, HGA tumor organoids derived from different patients demonstrated variable chemotherapy tumor-killing responses, whereas LGA organoids tested with the same regimens showed no response to chemotherapy. One LGA set of organoids was immune-enhanced with cells from a patient-matched lymph node to demonstrate feasibility of a symbiotic 3D reconstruction of a patient matched tumor and immune system component. CONCLUSIONS Development of 3D appendiceal tumor organoids is feasible even in low cellularity LGA tumors, allowing for individual patient tumors to remain viable for research and personalized drug screening.
Collapse
Affiliation(s)
- Konstantinos I Votanopoulos
- Department of Surgery - Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA. .,Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA.
| | - Andrea Mazzocchi
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hemamylammal Sivakumar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Steven Forsythe
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Julio Aleman
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Edward A Levine
- Department of Surgery - Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA. .,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
1235
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
1236
|
Korch C, Varella-Garcia M. Tackling the Human Cell Line and Tissue Misidentification Problem Is Needed for Reproducible Biomedical Research. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.yamp.2018.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
1237
|
Gao M, Lin M, Moffitt RA, Salazar MA, Park J, Vacirca J, Huang C, Shroyer KR, Choi M, Georgakis GV, Sasson AR, Talamini MA, Kim J. Direct therapeutic targeting of immune checkpoint PD-1 in pancreatic cancer. Br J Cancer 2018; 120:88-96. [PMID: 30377341 PMCID: PMC6325157 DOI: 10.1038/s41416-018-0298-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) hijacks innate cellular processes to promote cancer growth. We hypothesized that PC exploits PD-1/PD-L1 not only to avoid immune responses, but to directly enhance growth. We also hypothesized that immune checkpoint inhibitors (ICIs) have direct cytotoxicity in PC. We sought to elucidate therapeutic targeting of PD-1/PD-L1. METHODS PD-1 was assessed in PC cells, patient-derived organoids (PDOs), and clinical tissues. Then, PC cells were exposed to PD-L1 to evaluate proliferation. To test PD-1/PD-L1 signaling, cells were exposed to PD-L1 and MAPK was examined. Radio-immunoconjugates with anti-PD-1 drugs were developed to test uptake in patient-derived tumor xenografts (PDTXs). Next, PD-1 function was assessed by xenografting PD-1-knockdown cells. Finally, PC models were exposed to ICIs. RESULTS PD-1 expression was demonstrated in PCs. PD-L1 exposure increased proliferation and activated MAPK. Imaging PDTXs revealed uptake of radio-immunoconjugates. PD-1 knockdown in vivo revealed 67% smaller volumes than controls. Finally, ICI treatment of both PDOs/PDTXs demonstrated cytotoxicity and anti-MEK1/2 combined with anti-PD-1 drugs produced highest cytotoxicity in PDOs/PDTXs. CONCLUSIONS Our data reveal PCs innately express PD-1 and activate druggable oncogenic pathways supporting PDAC growth. Strategies directly targeting PC with novel ICI regimens may work with adaptive immune responses for optimal cytotoxicity.
Collapse
Affiliation(s)
- Mei Gao
- Department of Surgery, University of Kentucky, Lexington, KY, USA.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Miranda Lin
- Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Richard A Moffitt
- Department of Pathology, State University of New York, Stony Brook, NY, USA
| | - Marcela A Salazar
- Department of Experimental Therapeutics, City of Hope, Duarte, CA, USA
| | - Jinha Park
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Jeffrey Vacirca
- New York Cancer Specialists, East Setauket, New York, NY, USA
| | - Chuan Huang
- Departments of Radiology, State University of New York, Stony Brook, NY, USA.,Departments of Psychiatry, State University of New York, Stony Brook, NY, USA
| | - Kenneth R Shroyer
- Department of Pathology, State University of New York, Stony Brook, NY, USA
| | - Minsig Choi
- Departments of Medicine, State University of New York, Stony Brook, NY, USA
| | | | - Aaron R Sasson
- Departments of Surgery, State University of New York, Stony Brook, NY, USA
| | - Mark A Talamini
- Departments of Surgery, State University of New York, Stony Brook, NY, USA
| | - Joseph Kim
- Department of Surgery, University of Kentucky, Lexington, KY, USA. .,Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
1238
|
Castellón EA. Patient-derived organoids: New co-clinical model to predict treatment response in cancer? Oral Dis 2018; 25:928-930. [PMID: 30281877 DOI: 10.1111/odi.12988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Enrique A Castellón
- Department of Basic and Clinic Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
1239
|
Tanaka N, Osman AA, Takahashi Y, Lindemann A, Patel AA, Zhao M, Takahashi H, Myers JN. Head and neck cancer organoids established by modification of the CTOS method can be used to predict in vivo drug sensitivity. Oral Oncol 2018; 87:49-57. [PMID: 30527243 DOI: 10.1016/j.oraloncology.2018.10.018] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Currently there are no standard biomarkers of head and neck squamous cell carcinoma (HNSCC) response to therapy. This is, due to a lack of adequate predictive tumor models. To this end, we established cancer organoid lines from individual patient's tumors, and characterized their growth characteristics and response to different drug treatments with the objective of using these models for prediction of treatment response. MATERIALS AND METHODS Forty-three patients' samples were processed to establish organoids. To analyze the character of these organoids, immunohistochemistry, Western blotting, drug sensitivity assays, clonogenic survival assays, and animal experiments were performed. The HPV status and TP53 mutational status were also confirmed in these lines. RESULTS HNSCC organoids were successfully established with success rate of 30.2%. Corresponding two-dimensional cell lines were established from HNSCC organoids at higher success rate (53.8%). These organoids showed similar histological features and stem cell, epithelial and mesenchymal marker expression to the original tumors, thus recapitulating many of the characteristics of the original tumor cells. The cisplatin and docetaxel IC50 were determined for HNSCC organoids and the corresponding 2D cell lines using drug sensitivity and clonogenic survival assays. Responses to drug treatment in vivo were found to be similar to the IC50 calculated from organoids by drug sensitivity assays in vitro. CONCLUSION We established novel in vitro HNSCC cancer organoid lines retaining many properties of the original tumors from they were derived. These organoids can predict in vivo drug sensitivity and may represent useful tools to develop precision treatments for HNSCC.
Collapse
Affiliation(s)
- Noriaki Tanaka
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yoko Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Antje Lindemann
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ameeta A Patel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hideaki Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
1240
|
A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem Cell 2018; 23:882-897.e11. [PMID: 30344100 DOI: 10.1016/j.stem.2018.09.016] [Citation(s) in RCA: 473] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/20/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022]
Abstract
Gastric cancer displays marked molecular heterogeneity with aggressive behavior and treatment resistance. Therefore, good in vitro models that encompass unique subtypes are urgently needed for precision medicine development. Here, we have established a primary gastric cancer organoid (GCO) biobank that comprises normal, dysplastic, cancer, and lymph node metastases (n = 63) from 34 patients, including detailed whole-exome and transcriptome analysis. The cohort encompasses most known molecular subtypes (including EBV, MSI, intestinal/CIN, and diffuse/GS, with CLDN18-ARHGAP6 or CTNND1-ARHGAP26 fusions or RHOA mutations), capturing regional heterogeneity and subclonal architecture, while their morphology, transcriptome, and genomic profiles remain closely similar to in vivo tumors, even after long-term culture. Large-scale drug screening revealed sensitivity to unexpected drugs that were recently approved or in clinical trials, including Napabucasin, Abemaciclib, and the ATR inhibitor VE-822. Overall, this new GCO biobank, with linked genomic data, provides a useful resource for studying both cancer cell biology and precision cancer therapy.
Collapse
|
1241
|
Yang H, Sun L, Liu M, Mao Y. Patient-derived organoids: a promising model for personalized cancer treatment. Gastroenterol Rep (Oxf) 2018; 6:243-245. [PMID: 30430011 PMCID: PMC6225812 DOI: 10.1093/gastro/goy040] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/16/2018] [Accepted: 10/09/2018] [Indexed: 01/03/2023] Open
Affiliation(s)
- Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Meixi Liu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, P.R. China
| |
Collapse
|
1242
|
Su QP, Ju LA. Biophysical nanotools for single-molecule dynamics. Biophys Rev 2018; 10:1349-1357. [PMID: 30121743 PMCID: PMC6233351 DOI: 10.1007/s12551-018-0447-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022] Open
Abstract
The focus of the cell biology field is now shifting from characterizing cellular activities to organelle and molecular behaviors. This process accompanies the development of new biophysical visualization techniques that offer high spatial and temporal resolutions with ultra-sensitivity and low cell toxicity. They allow the biology research community to observe dynamic behaviors from scales of single molecules, organelles, cells to organoids, and even live animal tissues. In this review, we summarize these biophysical techniques into two major classes: the mechanical nanotools like dynamic force spectroscopy (DFS) and the optical nanotools like single-molecule and super-resolution microscopy. We also discuss their applications in elucidating molecular dynamics and functionally mapping of interactions between inter-cellular networks and intra-cellular components, which is key to understanding cellular processes such as adhesion, trafficking, inheritance, and division.
Collapse
Affiliation(s)
- Qian Peter Su
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, 2007, Australia.
| | - Lining Arnold Ju
- Charles Perkins Centre and Heart Research Institute, University of Sydney, Camperdown, New South Wales, 2006, Australia.
| |
Collapse
|
1243
|
|
1244
|
Khan KH, Cunningham D, Werner B, Vlachogiannis G, Spiteri I, Heide T, Mateos JF, Vatsiou A, Lampis A, Damavandi MD, Lote H, Huntingford IS, Hedayat S, Chau I, Tunariu N, Mentrasti G, Trevisani F, Rao S, Anandappa G, Watkins D, Starling N, Thomas J, Peckitt C, Khan N, Rugge M, Begum R, Hezelova B, Bryant A, Jones T, Proszek P, Fassan M, Hahne JC, Hubank M, Braconi C, Sottoriva A, Valeri N. Longitudinal Liquid Biopsy and Mathematical Modeling of Clonal Evolution Forecast Time to Treatment Failure in the PROSPECT-C Phase II Colorectal Cancer Clinical Trial. Cancer Discov 2018; 8:1270-1285. [PMID: 30166348 PMCID: PMC6380469 DOI: 10.1158/2159-8290.cd-17-0891] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 05/01/2018] [Accepted: 07/05/2018] [Indexed: 12/14/2022]
Abstract
Sequential profiling of plasma cell-free DNA (cfDNA) holds immense promise for early detection of patient progression. However, how to exploit the predictive power of cfDNA as a liquid biopsy in the clinic remains unclear. RAS pathway aberrations can be tracked in cfDNA to monitor resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. In this prospective phase II clinical trial of single-agent cetuximab in RAS wild-type patients, we combine genomic profiling of serial cfDNA and matched sequential tissue biopsies with imaging and mathematical modeling of cancer evolution. We show that a significant proportion of patients defined as RAS wild-type based on diagnostic tissue analysis harbor aberrations in the RAS pathway in pretreatment cfDNA and, in fact, do not benefit from EGFR inhibition. We demonstrate that primary and acquired resistance to cetuximab are often of polyclonal nature, and these dynamics can be observed in tissue and plasma. Furthermore, evolutionary modeling combined with frequent serial sampling of cfDNA allows prediction of the expected time to treatment failure in individual patients. This study demonstrates how integrating frequently sampled longitudinal liquid biopsies with a mathematical framework of tumor evolution allows individualized quantitative forecasting of progression, providing novel opportunities for adaptive personalized therapies.Significance: Liquid biopsies capture spatial and temporal heterogeneity underpinning resistance to anti-EGFR monoclonal antibodies in colorectal cancer. Dense serial sampling is needed to predict the time to treatment failure and generate a window of opportunity for intervention. Cancer Discov; 8(10); 1270-85. ©2018 AACR. See related commentary by Siravegna and Corcoran, p. 1213 This article is highlighted in the In This Issue feature, p. 1195.
Collapse
Affiliation(s)
- Khurum H Khan
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Benjamin Werner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Georgios Vlachogiannis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Timon Heide
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Javier Fernandez Mateos
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Alexandra Vatsiou
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Mahnaz Darvish Damavandi
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Hazel Lote
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Said Huntingford
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Somaieh Hedayat
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Giulia Mentrasti
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Francesco Trevisani
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Gayathri Anandappa
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Janet Thomas
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Clare Peckitt
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nasir Khan
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Massimo Rugge
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Blanka Hezelova
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Annette Bryant
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Thomas Jones
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Paula Proszek
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Matteo Fassan
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Jens C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Michael Hubank
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom.
| | - Nicola Valeri
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom.
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| |
Collapse
|
1245
|
Broguiere N, Isenmann L, Hirt C, Ringel T, Placzek S, Cavalli E, Ringnalda F, Villiger L, Züllig R, Lehmann R, Rogler G, Heim MH, Schüler J, Zenobi-Wong M, Schwank G. Growth of Epithelial Organoids in a Defined Hydrogel. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801621. [PMID: 30203567 DOI: 10.1002/adma.201801621] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/11/2018] [Indexed: 05/21/2023]
Abstract
Epithelial organoids are simplified models of organs grown in vitro from embryonic and adult stem cells. They are widely used to study organ development and disease, and enable drug screening in patient-derived primary tissues. Current protocols, however, rely on animal- and tumor-derived basement membrane extract (BME) as a 3D scaffold, which limits possible applications in regenerative medicine. This prompted us to study how organoids interact with their matrix, and to develop a well-defined hydrogel that supports organoid generation and growth. It is found that soft fibrin matrices provide suitable physical support, and that naturally occurring Arg-Gly-Asp (RGD) adhesion domains on the scaffold, as well as supplementation with laminin-111, are key parameters required for robust organoid formation and expansion. The possibility to functionalize fibrin via factor XIII-mediated anchoring also allows to covalently link fluorescent nanoparticles to the matrix for 3D traction force microscopy. These measurements suggest that the morphogenesis of budding intestinal organoids results from internal pressure combined with higher cell contractility in the regions containing differentiated cells compared to the regions containing stem cells. Since the fibrin/laminin matrix supports long-term expansion of all tested murine and human epithelial organoids, this hydrogel can be widely used as a defined equivalent to BME.
Collapse
Affiliation(s)
- Nicolas Broguiere
- Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Luca Isenmann
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Christian Hirt
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Till Ringel
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Silja Placzek
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Emma Cavalli
- Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Femke Ringnalda
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Lukas Villiger
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Richard Züllig
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital, Zurich, 8091, Switzerland
| | - Roger Lehmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital, Zurich, 8091, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, CH-4031, Switzerland
- Division of Gastroenterology and Hepatology, University Hospital Basel, Basel, CH-4031, Switzerland
| | - Julia Schüler
- Charles River Research Services Germany GmbH, Am Flughafen 12, 79108, Freiburg im Breisgau, Germany
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Gerald Schwank
- Institute of Molecular Health Sciences, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| |
Collapse
|
1246
|
|
1247
|
Clinton J, McWilliams-Koeppen P. Initiation, Expansion, and Cryopreservation of Human Primary Tissue-Derived Normal and Diseased Organoids in Embedded Three-Dimensional Culture. ACTA ACUST UNITED AC 2018; 82:e66. [PMID: 30265443 DOI: 10.1002/cpcb.66] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Organoids are primary patient-derived micro tissues grown within a three-dimensional extracellular matrix that better represents in vivo physiology and genetic diversity than existing two-dimensional cell lines. Organoids rely on the self-renewal and differentiation of tissue-resident stem cells that expand in culture and self-organize into complex three-dimensional structures. Depending on the tissue, organoids typically lack stromal, vascular, neural, and immune cells but otherwise can contain cells from all the respective tissue-specific cell lineages found in vivo. Established organoids can be initiated from cryopreserved material, cultured using largely traditional cell culture techniques and equipment, and then expanded and cryopreserved for future use. Organoid models have been developed from a variety of diseased and normal tissues including small intestine, colon, mammary, esophagus, lung, prostate, and pancreas. © 2018 by John Wiley & Sons, Inc.
Collapse
|
1248
|
Kim H, Schaniel C. Modeling Hematological Diseases and Cancer With Patient-Specific Induced Pluripotent Stem Cells. Front Immunol 2018; 9:2243. [PMID: 30323816 PMCID: PMC6172418 DOI: 10.3389/fimmu.2018.02243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) together with recent advances in genome editing, microphysiological systems, tissue engineering and xenograft models present new opportunities for the investigation of hematological diseases and cancer in a patient-specific context. Here we review the progress in the field and discuss the advantages, limitations, and challenges of iPSC-based malignancy modeling. We will also discuss the use of iPSCs and its derivatives as cellular sources for drug target identification, drug development and evaluation of pharmacological responses.
Collapse
Affiliation(s)
- Huensuk Kim
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
1249
|
Zhang F, Wang W, Long Y, Liu H, Cheng J, Guo L, Li R, Meng C, Yu S, Zhao Q, Lu S, Wang L, Wang H, Wen D. Characterization of drug responses of mini patient-derived xenografts in mice for predicting cancer patient clinical therapeutic response. Cancer Commun (Lond) 2018; 38:60. [PMID: 30257718 PMCID: PMC6158900 DOI: 10.1186/s40880-018-0329-5] [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] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/15/2018] [Indexed: 12/24/2022] Open
Abstract
Background Patient-derived organoids and xenografts (PDXs) have emerged as powerful models in functional diagnostics with high predictive power for anticancer drug response. However, limitations such as engraftment failure and time-consuming for establishing and expanding PDX models followed by testing drug efficacy, and inability to subject to systemic drug administration for ex vivo organoid culture hinder realistic and fast decision-making in selecting the right therapeutics in the clinic. The present study aimed to develop an advanced PDX model, namely MiniPDX, for rapidly testing drug efficacy to strengthen its value in personalized cancer treatment. Methods We developed a rapid in vivo drug sensitivity assay, OncoVee® MiniPDX, for screening clinically relevant regimens for cancer. In this model, patient-derived tumor cells were arrayed within hollow fiber capsules, implanted subcutaneously into mice and cultured for 7 days. The cellular activity morphology and pharmacokinetics were systematically evaluated. MiniPDX performance (sensitivity, specificity, positive and negative predictive values) was examined using PDX as the reference. Drug responses were examined by tumor cell growth inhibition rate and tumor growth inhibition rate in PDX models and MiniPDX assays respectively. The results from MiniPDX were also used to evaluate its predictive power for clinical outcomes. Results Morphological and histopathological features of tumor cells within the MiniPDX capsules matched those both in PDX models and in original tumors. Drug responses in the PDX tumor graft assays correlated well with those in the corresponding MiniPDX assays using 26 PDX models generated from patients, including 14 gastric cancer, 10 lung cancer and 2 pancreatic cancer. The positive predictive value of MiniPDX was 92%, and the negative predictive value was 81% with a sensitivity of 80% and a specificity of 93%. Through expanding to clinical tumor samples, MiniPDX assay showed potential of wide clinical application. Conclusions Fast in vivo MiniPDX assay based on capsule implantation was developed-to assess drug responses of both PDX tumor grafts and clinical cancer specimens. The high correlation between drug responses of paired MiniPDX and PDX tumor graft assay, as well as translational data suggest that MiniPDX assay is an advanced tool for personalized cancer treatment.
Collapse
Affiliation(s)
- Feifei Zhang
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Wenjie Wang
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Yuan Long
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Hui Liu
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Jijun Cheng
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Lin Guo
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Rongyu Li
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Chao Meng
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Shan Yu
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China
| | - Qingchuan Zhao
- Department of Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Shun Lu
- Department of Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Lili Wang
- The Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Urology, Tianjin, 300211, P. R. China
| | - Haitao Wang
- The Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Urology, Tianjin, 300211, P. R. China
| | - Danyi Wen
- Shanghai LIDE Biotech Co., LTD, Shanghai, 201203, P. R. China.
| |
Collapse
|
1250
|
Abstract
Over the past six decades the inflation-adjusted cost to bring a new drug to market has been increasing constantly and doubles every 9 years - now reaching in excess of $2.5 billion. Overall, the likelihood of FDA approval for a drug (any disease indication) that has entered phase I clinical trials is a mere 9.6%, with the approval rate for oncology far below average at only 5.1%. Lack of efficacy or toxicity is often not revealed until the later stages of clinical trials, despite promising preclinical data. This indicates that the current in vitro systems for drug screening need to be improved for better predictability of in vivo outcomes. Microphysiological systems (MPS), or bioengineered 3D microfluidic tissue and organ constructs that mimic physiological and pathological processes in vitro, can be leveraged across preclinical research and clinical trial stages to transform drug development and clinical management for a range of diseases. Here we review the current state-of-the-art in 3D tissue-engineering models developed for cancer research, with a focus on tumor-on-a-chip, or tumor chip, models. From our viewpoint, tumor chip systems can advance innovative medicine to ameliorate the high failure rates in anti-cancer drug development and clinical treatment.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697, USA.
| | | |
Collapse
|