1
|
Bastian JLD, Zeuschner P, Stöckle M, Junker K, Linxweiler J. Tumor promoting effect of spheroids in an orthotopic prostate cancer mouse model. Sci Rep 2024; 14:8835. [PMID: 38632341 PMCID: PMC11024136 DOI: 10.1038/s41598-024-59052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
In this study, we aimed to establish a technique for intraprostatic implantation of prostate cancer (PCa) spheroids and to identify the impact of three-dimensional organization of PCa cells on tumor progression and metastasis in a representative in vivo model. 40,000 LNCaP cells were implanted into the prostate of immunodeficient SCID mice either as single cells (n = 8) or as preformed 3D spheroids (n = 8). For a follow up of 20 weeks, tumor growth was monitored by serum PSA and high-resolution 3D ultrasonography. Eventually, animals were sacrificed and autopsied. The organ dissects were analyzed for the presence of metastases by histology (H&E) and immunohistochemistry (AMACR, AR, Ki-67, CK5, CK8, E-Cadherin, Vimentin). Solid intraprostatic tumors developed in 50% of mice after spheroid implantation and in 50% of mice after implantation of a single cells. Primary tumors of LNCaP spheroids evolved earlier, exhibiting a shorter tumor doubling time whilst developing larger tumor volumes, which was reflected by a higher immunohistochemical expression of Ki-67 and AR, too. Spheroid tumors established lung and lymph node metastases in 75% of mice, in contrast to 50% of mice after single cell implantation. Our technique enables a variety of studies regarding the influence of the tumor microenvironment on PCa progression.
Collapse
Affiliation(s)
- Julius Lars Daniel Bastian
- Department of Urology and Pediatric Urology, Saarland University, Kirrbergerstr. 100 Gebäude 6, 66424, Homburg, Germany
| | - Philip Zeuschner
- Department of Urology and Pediatric Urology, Saarland University, Kirrbergerstr. 100 Gebäude 6, 66424, Homburg, Germany
| | - Michael Stöckle
- Department of Urology and Pediatric Urology, Saarland University, Kirrbergerstr. 100 Gebäude 6, 66424, Homburg, Germany
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Kirrbergerstr. 100 Gebäude 6, 66424, Homburg, Germany
| | - Johannes Linxweiler
- Department of Urology and Pediatric Urology, Saarland University, Kirrbergerstr. 100 Gebäude 6, 66424, Homburg, Germany.
| |
Collapse
|
2
|
Paindelli C, Parietti V, Barrios S, Shepherd P, Pan T, Wang WL, Satcher RL, Logothetis CJ, Navone N, Campbell MT, Mikos AG, Dondossola E. Bone mimetic environments support engineering, propagation, and analysis of therapeutic response of patient-derived cells, ex vivo and in vivo. Acta Biomater 2024; 178:83-92. [PMID: 38387748 DOI: 10.1016/j.actbio.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/22/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Bone metastases are the most common milestone in the lethal progression of prostate cancer and prominent in a substantial portion of renal malignancies. Interactions between cancer and bone host cells have emerged as drivers of both disease progression and therapeutic resistance. To best understand these central host-epithelial cell interactions, biologically relevant preclinical models are required. To achieve this goal, we here established and characterized tissue-engineered bone mimetic environments (BME) capable of supporting the growth of patient-derived xenograft (PDX) cells, ex vivo and in vivo. The BME consisted of a polycaprolactone (PCL) scaffold colonized by human mesenchymal stem cells (hMSCs) differentiated into osteoblasts. PDX-derived cells were isolated from bone metastatic prostate or renal tumors, engineered to express GFP or luciferase and seeded onto the BMEs. BMEs supported the growth and therapy response of PDX-derived cells, ex vivo. Additionally, BMEs survived after in vivo implantation and further sustained the growth of PDX-derived cells, their serial transplant, and their application to study the response to treatment. Taken together, this demonstrates the utility of BMEs in combination with patient-derived cells, both ex vivo and in vivo. STATEMENT OF SIGNIFICANCE: Our tissue-engineered BME supported the growth of patient-derived cells and proved useful to monitor the therapy response, both ex vivo and in vivo. This approach has the potential to enable co-clinical strategies to monitor bone metastatic tumor progression and therapy response, including identification and prioritization of new targets for patient treatment.
Collapse
Affiliation(s)
- Claudia Paindelli
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Vanessa Parietti
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States; Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Peter Shepherd
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Tianhong Pan
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Robert L Satcher
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Nora Navone
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Antonios G Mikos
- Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States.
| |
Collapse
|
3
|
Liu AY. Prostate cancer research: tools, cell types, and molecular targets. Front Oncol 2024; 14:1321694. [PMID: 38595814 PMCID: PMC11002103 DOI: 10.3389/fonc.2024.1321694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Multiple cancer cell types are found in prostate tumors. They are either luminal-like adenocarcinoma or less luminal-like and more stem-like non-adenocarcinoma and small cell carcinoma. These types are lineage related through differentiation. Loss of cancer differentiation from luminal-like to stem-like is mediated by the activation of stem cell transcription factors (scTF) such as LIN28A, NANOG, POU5F1 and SOX2. scTF expression leads to down-regulation of β2-microglobulin (B2M). Thus, cancer cells can change from the scT F ˜ B 2 M hi phenotype of differentiated to that of scT F ˙ B 2 M lo of dedifferentiated in the disease course. In development, epithelial cell differentiation is induced by stromal signaling and cell contact. One of the stromal factors specific to prostate encodes proenkephalin (PENK). PENK can down-regulate scTF and up-regulate B2M in stem-like small cell carcinoma LuCaP 145.1 cells indicative of exit from the stem state and differentiation. In fact, prostate cancer cells can be made to undergo dedifferentiation or reprogramming by scTF transfection and then to differentiate by PENK transfection. Therapies need to be designed for treating the different cancer cell types. Extracellular anterior gradient 2 (eAGR2) is an adenocarcinoma antigen associated with cancer differentiation that can be targeted by antibodies to lyse tumor cells with immune system components. eAGR2 is specific to cancer as normal cells express only the intracellular form (iAGR2). For AGR2-negative stem-like cancer cells, factors like PENK that can target scTF could be effective in differentiation therapy.
Collapse
Affiliation(s)
- Alvin Y. Liu
- Department of Urology, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
4
|
Thomas PB, Alinezhad S, Joshi A, Sweeney K, Tse BWC, Tevz G, McPherson S, Nelson CC, Williams ED, Vela I. Introduction of Androgen Receptor Targeting shRNA Inhibits Tumor Growth in Patient-Derived Prostate Cancer Xenografts. Curr Oncol 2023; 30:9437-9447. [PMID: 37999103 PMCID: PMC10670201 DOI: 10.3390/curroncol30110683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/16/2023] [Accepted: 10/21/2023] [Indexed: 11/25/2023] Open
Abstract
Patient-derived xenograft (PDX) models have been established as important preclinical cancer models, overcoming some of the limitations associated with the use of cancer cell lines. The utility of prostate cancer PDX models has been limited by an inability to genetically manipulate them in vivo and difficulties sustaining PDX-derived cancer cells in culture. Viable, short-term propagation of PDX models would allow in vitro transfection with traceable reporters or manipulation of gene expression relevant to different studies within the prostate cancer field. Here, we report an organoid culture system that supports the growth of prostate cancer PDX cells in vitro and permits genetic manipulation, substantially increasing the scope to use PDXs to study the pathobiology of prostate cancer and define potential therapeutic targets. We have established a short-term PDX-derived in vitro cell culture system which enables genetic manipulation of prostate cancer PDXs LuCaP35 and BM18. Genetically manipulated cells could be re-established as viable xenografts when re-implanted subcutaneously in immunocompromised mice and were able to be serially passaged. Tumor growth of the androgen-dependent LuCaP35 PDX was significantly inhibited following depletion of the androgen receptor (AR) in vivo. Taken together, this system provides a method to generate novel preclinical models to assess the impact of controlled genetic perturbations and allows for targeting specific genes of interest in the complex biological setting of solid tumors.
Collapse
Affiliation(s)
- Patrick B. Thomas
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD 4102, Australia
| | - Saeid Alinezhad
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
| | - Andre Joshi
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Katrina Sweeney
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
| | - Brian W. C. Tse
- Preclinical Imaging Facility, Translational Research Institute (TRI), Brisbane, QLD 4102, Australia;
| | - Gregor Tevz
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
| | - Stephen McPherson
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
| | - Colleen C. Nelson
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Elizabeth D. Williams
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD 4102, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Ian Vela
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.)
- Australian Prostate Cancer Research Centre—Queensland, Brisbane, QLD 4102, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| |
Collapse
|
5
|
Sailer V, von Amsberg G, Duensing S, Kirfel J, Lieb V, Metzger E, Offermann A, Pantel K, Schuele R, Taubert H, Wach S, Perner S, Werner S, Aigner A. Experimental in vitro, ex vivo and in vivo models in prostate cancer research. Nat Rev Urol 2023; 20:158-178. [PMID: 36451039 DOI: 10.1038/s41585-022-00677-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/02/2022]
Abstract
Androgen deprivation therapy has a central role in the treatment of advanced prostate cancer, often causing initial tumour remission before increasing independence from signal transduction mechanisms of the androgen receptor and then eventual disease progression. Novel treatment approaches are urgently needed, but only a fraction of promising drug candidates from the laboratory will eventually reach clinical approval, highlighting the demand for critical assessment of current preclinical models. Such models include standard, genetically modified and patient-derived cell lines, spheroid and organoid culture models, scaffold and hydrogel cultures, tissue slices, tumour xenograft models, patient-derived xenograft and circulating tumour cell eXplant models as well as transgenic and knockout mouse models. These models need to account for inter-patient and intra-patient heterogeneity, the acquisition of primary or secondary resistance, the interaction of tumour cells with their microenvironment, which make crucial contributions to tumour progression and resistance, as well as the effects of the 3D tissue network on drug penetration, bioavailability and efficacy.
Collapse
Affiliation(s)
- Verena Sailer
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Gunhild von Amsberg
- Department of Oncology and Hematology, University Cancer Center Hamburg Eppendorf and Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg and National Center for Tumour Diseases, Heidelberg, Germany
| | - Jutta Kirfel
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Verena Lieb
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Eric Metzger
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Anne Offermann
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Klaus Pantel
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Roland Schuele
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Helge Taubert
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Wach
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Perner
- University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Stefan Werner
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Achim Aigner
- Clinical Pharmacology, Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Medical Faculty, Leipzig, Germany.
| |
Collapse
|
6
|
Geometric tumor embolic budding characterizes inflammatory breast cancer. Breast Cancer Res Treat 2023; 197:461-478. [PMID: 36473978 PMCID: PMC9734724 DOI: 10.1007/s10549-022-06819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE Inflammatory breast cancer (IBC) is characterized by numerous tumor emboli especially within dermal lymphatics. The explanation remains a mystery. METHODS This study combines experimental studies with two different IBC xenografts with image algorithmic studies utilizing human tissue microarrays (TMAs) of IBC vs non-IBC cases to support a novel hypothesis to explain IBC's sina qua non signature of florid lymphovascular emboli. RESULTS In the human TMAs, compared to tumor features like nuclear grade (size), mitosis and Ki-67 immunoreactivity which show that IBC is only modestly more proliferative with larger nuclei than non-IBC, what really sets IBC apart is the markedly greater number of tumor emboli and distinctly smaller emboli whose numbers indicate geometric or exponential differences between IBC and non-IBC. In the experimental xenograft studies, Mary-X gives rise to tight spheroids in vitro which exhibit dynamic budding into smaller daughter spheroids whereas Karen-X exhibits only loose non-budding aggregates. Furthermore Mary-X emboli also bud dramatically into smaller daughter emboli in vivo. The mechanism that regulates this involves the generation of E-cad/NTF1, a calpain-mediated cleavage 100 kDa product of 120 kDa full length membrane E-cadherin. Inhibiting this calpain-mediated cleavage of E-cadherin by blocking either the calpain site of cleavage (SC) or the site of binding (SB) with specific decapeptides that both penetrate the cell membrane and mimic either the cleavage site or the binding site on E-cadherin, inhibits the generation of E-cad/NTF1 in a dose-dependent manner, reduces spheroid compactness and decreases budding. CONCLUSION Since E-cad/NFT1 retains the p120ctn binding site but loses the α-and β-catenin sites, promoting its 360° distribution around the cell's membrane, the vacilating levels of this molecule trigger budding of both the spheroids as well as the emboli. Recurrent and geometric budding of parental emboli into daughter emboli then would account for the plethora of emboli seen in IBC.
Collapse
|
7
|
Rice MA, Kumar V, Tailor D, Garcia-Marques FJ, Hsu EC, Liu S, Bermudez A, Kanchustambham V, Shankar V, Inde Z, Alabi BR, Muruganantham A, Shen M, Pandrala M, Nolley R, Aslan M, Ghoochani A, Agarwal A, Buckup M, Kumar M, Going CC, Peehl DM, Dixon SJ, Zare RN, Brooks JD, Pitteri SJ, Malhotra SV, Stoyanova T. SU086, an inhibitor of HSP90, impairs glycolysis and represents a treatment strategy for advanced prostate cancer. Cell Rep Med 2022; 3:100502. [PMID: 35243415 PMCID: PMC8861828 DOI: 10.1016/j.xcrm.2021.100502] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/09/2021] [Accepted: 12/20/2021] [Indexed: 12/19/2022]
Abstract
Among men, prostate cancer is the second leading cause of cancer-associated mortality, with advanced disease remaining a major clinical challenge. We describe a small molecule, SU086, as a therapeutic strategy for advanced prostate cancer. We demonstrate that SU086 inhibits the growth of prostate cancer cells in vitro, cell-line and patient-derived xenografts in vivo, and ex vivo prostate cancer patient specimens. Furthermore, SU086 in combination with standard of care second-generation anti-androgen therapies displays increased impairment of prostate cancer cell and tumor growth in vitro and in vivo. Cellular thermal shift assay reveals that SU086 binds to heat shock protein 90 (HSP90) and leads to a decrease in HSP90 levels. Proteomic profiling demonstrates that SU086 binds to and decreases HSP90. Metabolomic profiling reveals that SU086 leads to perturbation of glycolysis. Our study identifies SU086 as a treatment for advanced prostate cancer as a single agent or when combined with second-generation anti-androgens.
Collapse
Affiliation(s)
- Meghan A. Rice
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Vineet Kumar
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Dhanir Tailor
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Fernando Jose Garcia-Marques
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - En-Chi Hsu
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Shiqin Liu
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Abel Bermudez
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | | | - Vishnu Shankar
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Zintis Inde
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Busola Ruth Alabi
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Arvind Muruganantham
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Michelle Shen
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Mallesh Pandrala
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rosalie Nolley
- Department of Urology, Stanford University, Stanford, CA 94305, USA
| | - Merve Aslan
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Ali Ghoochani
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Arushi Agarwal
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Mark Buckup
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Manoj Kumar
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Catherine C. Going
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Donna M. Peehl
- Department of Urology, Stanford University, Stanford, CA 94305, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott J. Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - James D. Brooks
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
- Department of Urology, Stanford University, Stanford, CA 94305, USA
| | - Sharon J. Pitteri
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| | - Sanjay V. Malhotra
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tanya Stoyanova
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Linxweiler J, Hajili T, Zeuschner P, Menger MD, Stöckle M, Junker K, Saar M. Primary Tumor Resection Decelerates Disease Progression in an Orthotopic Mouse Model of Metastatic Prostate Cancer. Cancers (Basel) 2022; 14:cancers14030737. [PMID: 35159004 PMCID: PMC8833735 DOI: 10.3390/cancers14030737] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 01/25/2023] Open
Abstract
Radical prostatectomy in oligometastatic prostate cancer is a matter of intense debate. Besides avoiding local complications, it is hypothesized that primary tumor resection may result in better oncological outcomes. The aim of our study was to analyze the effect of primary tumor resection on disease progression in an orthotopic prostate cancer mouse model. First, the optimal time point for primary tumor resection, when metastases have already occurred, but the primary tumor is still resectable, was determined as 8 weeks after inoculation of 5 × 105 LuCaP136 cells. In a second in vivo experiment, 64 mice with metastatic prostate cancer were randomized into two groups, primary tumor resection or sham operation, and disease progression was followed up for 10 weeks. The technique of orthotopic primary tumor resection was successfully established. Compared with the sham operation group, mice with primary tumor resection showed a significantly longer survival (p < 0.001), a significantly slower PSA increase (p < 0.01), and a lower number of lung metastases (p = 0.073). In conclusion, primary tumor resection resulted in slower disease progression and longer survival in an orthotopic mouse model of metastatic prostate cancer. In future studies, this model will be used to unravel the molecular mechanisms of primary tumor/metastasis interaction in prostate cancer.
Collapse
Affiliation(s)
- Johannes Linxweiler
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
- Correspondence:
| | - Turkan Hajili
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
| | - Philip Zeuschner
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
| | - Michael D. Menger
- Institute for Clinical-Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany;
| | - Michael Stöckle
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
| | - Kerstin Junker
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
| | - Matthias Saar
- Department of Urology, Saarland University, 66421 Homburg, Saar, Germany; (T.H.); (P.Z.); (M.S.); (K.J.); (M.S.)
| |
Collapse
|
9
|
Ukidve A, Cu K, Kumbhojkar N, Lahann J, Mitragotri S. Overcoming biological barriers to improve solid tumor immunotherapy. Drug Deliv Transl Res 2021; 11:2276-2301. [PMID: 33611770 DOI: 10.1007/s13346-021-00923-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has been at the forefront of therapeutic interventions for many different tumor types over the last decade. While the discovery of immunotherapeutics continues to occur at an accelerated rate, their translation is often hindered by a lack of strategies to deliver them specifically into solid tumors. Accordingly, significant scientific efforts have been dedicated to understanding the underlying mechanisms that govern their delivery into tumors and the subsequent immune modulation. In this review, we aim to summarize the efforts focused on overcoming tumor-associated biological barriers and enhancing the potency of immunotherapy. We summarize the current understanding of biological barriers that limit the entry of intravascularly administered immunotherapies into the tumors, in vitro techniques developed to investigate the underlying transport processes, and delivery strategies developed to overcome the barriers. Overall, we aim to provide the reader with a framework that guides the rational development of technologies for improved solid tumor immunotherapy.
Collapse
Affiliation(s)
- Anvay Ukidve
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Katharina Cu
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Ninad Kumbhojkar
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Joerg Lahann
- Department of Chemical Engineering, Department of Material Science & Engineering, Department of Macromolecular Science & Engineering, Department of Biomedical Engineering, and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
11
|
Choo N, Ramm S, Luu J, Winter JM, Selth LA, Dwyer AR, Frydenberg M, Grummet J, Sandhu S, Hickey TE, Tilley WD, Taylor RA, Risbridger GP, Lawrence MG, Simpson KJ. High-Throughput Imaging Assay for Drug Screening of 3D Prostate Cancer Organoids. SLAS DISCOVERY 2021; 26:1107-1124. [PMID: 34111999 PMCID: PMC8458687 DOI: 10.1177/24725552211020668] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
New treatments are required for advanced prostate cancer; however, there are fewer preclinical models of prostate cancer than other common tumor types to test candidate therapeutics. One opportunity to increase the scope of preclinical studies is to grow tissue from patient-derived xenografts (PDXs) as organoid cultures. Here we report a scalable pipeline for automated seeding, treatment and an analysis of the drug responses of prostate cancer organoids. We established organoid cultures from 5 PDXs with diverse phenotypes of prostate cancer, including castrate-sensitive and castrate-resistant disease, as well as adenocarcinoma and neuroendocrine pathology. We robotically embedded organoids in Matrigel in 384-well plates and monitored growth via brightfield microscopy before treatment with poly ADP-ribose polymerase inhibitors or a compound library. Independent readouts including metabolic activity and live-cell imaging–based features provided robust measures of organoid growth and complementary ways of assessing drug efficacy. Single organoid analyses enabled in-depth assessment of morphological differences between patients and within organoid populations and revealed that larger organoids had more striking changes in morphology and composition after drug treatment. By increasing the scale and scope of organoid experiments, this automated assay complements other patient-derived models and will expedite preclinical testing of new treatments for prostate cancer.
Collapse
Affiliation(s)
- Nicholas Choo
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Susanne Ramm
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jennii Luu
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jean M Winter
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Amy R Dwyer
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Mark Frydenberg
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Department of Urology, Cabrini Health, Malvern, VIC, Australia
| | - Jeremy Grummet
- Australian Urology Associates, Melbourne, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Renea A Taylor
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Kaylene J Simpson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Daunys S, Janonienė A, Januškevičienė I, Paškevičiūtė M, Petrikaitė V. 3D Tumor Spheroid Models for In Vitro Therapeutic Screening of Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:243-270. [PMID: 33543463 DOI: 10.1007/978-3-030-58174-9_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The anticancer activity of compounds and nanoparticles is most often determined in the cell monolayer. However, three-dimensional (3D) systems, such as tumor spheroids, are more representing the natural tumor microenvironment. They have been shown to have higher invasiveness and resistance to cytotoxic agents and radiotherapy compared to cells growing in 2D monolayer. Furthermore, to improve the prediction of clinical efficacy of drugs, in the past decades, even more sophisticated systems, such as multicellular 3D cultures, closely representing natural tumor microenvironment have been developed. Those cultures are formed from either cell lines or patient-derived tumor cells. Such models are very attractive and could improve the selection of tested materials for clinical trials avoiding unnecessary expensive tests in vivo. The microenvironment in tumor spheroids is different, and those differences or the interaction between several cell populations may contribute to different tumor response to the treatment. Also, different types of nanoparticles may have different behavior in 3D models, depending on their nature, physicochemical properties, the presence of targeting ligands on the surface, etc. Therefore, it is very important to understand in which cases which type of tumor spheroid is more suitable for testing specific types of nanoparticles, which conditions should be used, and which analytical method should be applied.
Collapse
Affiliation(s)
- Simonas Daunys
- Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agnė Janonienė
- Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Indrė Januškevičienė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Miglė Paškevičiūtė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vilma Petrikaitė
- Life Sciences Center, Vilnius University, Vilnius, Lithuania.
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
- Institute of Physiology and Pharmacology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| |
Collapse
|
13
|
Linxweiler J, Hajili T, Körbel C, Berchem C, Zeuschner P, Müller A, Stöckle M, Menger MD, Junker K, Saar M. Cancer-associated fibroblasts stimulate primary tumor growth and metastatic spread in an orthotopic prostate cancer xenograft model. Sci Rep 2020; 10:12575. [PMID: 32724081 PMCID: PMC7387494 DOI: 10.1038/s41598-020-69424-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
The unique microenvironment of the prostate plays a crucial role in the development and progression of prostate cancer (PCa). We examined the effects of cancer-associated fibroblasts (CAFs) on PCa progression using patient-derived fibroblast primary cultures in a representative orthotopic xenograft model. Primary cultures of CAFs, non-cancer-associated fibroblasts (NCAFs) and benign prostate hyperplasia-associated fibroblasts (BPHFs) were generated from patient-derived tissue specimens. These fibroblasts were coinjected together with cancer cells (LuCaP136 spheroids or LNCaP cells) in orthotopic PCa xenografts to investigate their effects on local and systemic tumor progression. Primary tumor growth as well as metastatic spread to lymph nodes and lungs were significantly stimulated by CAF coinjection in LuCaP136 xenografts. When NCAFs or BPHFs were coinjected, tumor progression was similar to injection of tumor cells alone. In LNCaP xenografts, all three fibroblast types significantly stimulated primary tumor progression compared to injection of LNCaP cells alone. CAF coinjection further increased the frequency of lymph node and lung metastases. This is the first study using an orthotopic spheroid culture xenograft model to demonstrate a stimulatory effect of patient-derived CAFs on PCa progression. The established experimental setup will provide a valuable tool to further unravel the interacting mechanisms between PCa cells and their microenvironment.
Collapse
Affiliation(s)
- Johannes Linxweiler
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany.
| | - Turkan Hajili
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Carolina Berchem
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Philip Zeuschner
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Andreas Müller
- Department of Diagnostic and Interventional Radiology, Saarland University, Homburg/Saar, Germany
| | - Michael Stöckle
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Matthias Saar
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| |
Collapse
|
14
|
Joshi A, Roberts MJ, Alinezhad S, Williams ED, Vela I. Challenges, applications and future directions of precision medicine in prostate cancer - the role of organoids and patient-derived xenografts. BJU Int 2020; 126:65-72. [PMID: 32383524 DOI: 10.1111/bju.15103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To provide a clinically relevant outline of various current precision medicine principles and available evidence on the application and potential for a precision medicine approach in prostate cancer. METHODS Narrative review of the current literature in the field. CONCLUSION Precision medicine is the concept of individualising patient management based on specific tumour characteristics and biology, rather than traditional histological subtypes. The overall aim is to personalise management to individual patients, to provide the right cancer treatment, to the right patient, at the right time. While the approach aims to improve clinical outcomes, decrease morbidity and improve survival in men with advanced prostate cancer, its clinical application is in its infancy. It does however show great promise in this and other cancers, and will continue to be an area of active research and clinical investigation.
Collapse
Affiliation(s)
- Andre Joshi
- Department of Urology, Princess Alexandra Hospital, Brisbane, Qld, Australia.,Australian Prostate Cancer Research Centre-Queensland, Brisbane, Qld, Australia.,Translational Research Institute, Brisbane, Qld, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - Matthew J Roberts
- Department of Urology, Princess Alexandra Hospital, Brisbane, Qld, Australia.,University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Herston, Qld, Australia
| | - Saeid Alinezhad
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, Qld, Australia.,Translational Research Institute, Brisbane, Qld, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, Qld, Australia.,Translational Research Institute, Brisbane, Qld, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, Australia
| | - Ian Vela
- Department of Urology, Princess Alexandra Hospital, Brisbane, Qld, Australia.,Australian Prostate Cancer Research Centre-Queensland, Brisbane, Qld, Australia.,Translational Research Institute, Brisbane, Qld, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
15
|
Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G. Integrated cancer tissue engineering models for precision medicine. PLoS One 2019; 14:e0216564. [PMID: 31075118 PMCID: PMC6510431 DOI: 10.1371/journal.pone.0216564] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors are not merely cancerous cells that undergo mindless proliferation. Rather, they are highly organized and interconnected organ systems. Tumor cells reside in complex microenvironments in which they are subjected to a variety of physical and chemical stimuli that influence cell behavior and ultimately the progression and maintenance of the tumor. As cancer bioengineers, it is our responsibility to create physiologic models that enable accurate understanding of the multi-dimensional structure, organization, and complex relationships in diverse tumor microenvironments. Such models can greatly expedite clinical discovery and translation by closely replicating the physiological conditions while maintaining high tunability and control of extrinsic factors. In this review, we discuss the current models that target key aspects of the tumor microenvironment and their role in cancer progression. In order to address sources of experimental variation and model limitations, we also make recommendations for methods to improve overall physiologic reproducibility, experimental repeatability, and rigor within the field. Improvements can be made through an enhanced emphasis on mathematical modeling, standardized in vitro model characterization, transparent reporting of methodologies, and designing experiments with physiological metrics. Taken together these considerations will enhance the relevance of in vitro tumor models, biological understanding, and accelerate treatment exploration ultimately leading to improved clinical outcomes. Moreover, the development of robust, user-friendly models that integrate important stimuli will allow for the in-depth study of tumors as they undergo progression from non-transformed primary cells to metastatic disease and facilitate translation to a wide variety of biological and clinical studies.
Collapse
Affiliation(s)
- Michael E. Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric N. Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Caymen M. Novak
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine S. Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
16
|
Linxweiler J, Körbel C, Müller A, Hammer M, Veith C, Bohle RM, Stöckle M, Junker K, Menger MD, Saar M. A novel mouse model of human prostate cancer to study intraprostatic tumor growth and the development of lymph node metastases. Prostate 2018; 78:664-675. [PMID: 29572953 DOI: 10.1002/pros.23508] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/23/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND In this study, we aimed to establish a versatile in vivo model of prostate cancer, which adequately mimics intraprostatic tumor growth, and the natural routes of metastatic spread. In addition, we analyzed the capability of high-resolution ultrasonography (hrUS), in vivo micro-CT (μCT), and 9.4T MRI to monitor tumor growth and the development of lymph node metastases. METHODS A total of 5 × 105 VCaP cells or 5 × 105 cells of LuCaP136- or LuCaP147 spheroids were injected into the prostate of male CB17-SCID mice (n = 8 for each cell type). During 12 weeks of follow-up, orthotopic tumor growth, and metastatic spread were monitored by repetitive serum-PSA measurements and imaging studies including hrUS, μCT, and 9.4T MRI. At autopsy, primary tumors and metastases were harvested and examined by histology and immunohistochemistry (CK5, CK8, AMACR, AR, Ki67, ERG, and PSA). From imaging results and PSA-measurements, tumor volume doubling time, tumor-specific growth rate, and PSA-density were calculated. RESULTS All 24 mice developed orthotopic tumors. The tumor growth could be reliably monitored by a combination of hrUS, μCT, MRI, and serum-PSA measurements. In most animals, lymph node metastases could be detected after 12 weeks, which could also be well visualized by hrUS, and MRI. Immunohistochemistry showed positive signals for CK8, AMACR, and AR in all xenograft types. CK5 was negative in VCaP- and focally positive in LuCaP136- and LuCaP147-xenografts. ERG was positive in VCaP- and negative in LuCaP136- and LuCaP147-xenografts. Tumor volume doubling times and tumor-specific growth rates were 21.2 days and 3.9 %/day for VCaP-, 27.6 days and 3.1 %/day for LuCaP136- and 16.2 days and 4.5 %/day for LuCaP147-xenografts, respectively. PSA-densities were 433.9 ng/mL per milliliter tumor for VCaP-, 6.5 ng/mL per milliliter tumor for LuCaP136-, and 11.2 ng/mL per milliliter tumor for LuCaP147-xenografts. CONCLUSIONS By using different monolayer and 3D spheroid cell cultures in an orthotopic xenograft model, we established an innovative, versatile in vivo model of prostate cancer, which enables the study of both intraprostatic tumor growth as well as metastatic spread to regional lymph nodes. HrUS and MRI are feasible tools to monitor tumor growth and the development of lymph node metastases while these cannot be visualized by μCT.
Collapse
Affiliation(s)
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Andreas Müller
- Department of Diagnostic and Interventional Radiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Markus Hammer
- Department of Urology, Saarland University, Homburg/Saar, Germany
| | - Christian Veith
- Department of General and Surgical Pathology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Rainer M Bohle
- Department of General and Surgical Pathology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Michael Stöckle
- Department of Urology, Saarland University, Homburg/Saar, Germany
| | - Kerstin Junker
- Department of Urology, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Saar
- Department of Urology, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
17
|
Kohli M, Ho Y, Hillman DW, Van Etten JL, Henzler C, Yang R, Sperger JM, Li Y, Tseng E, Hon T, Clark T, Tan W, Carlson RE, Wang L, Sicotte H, Thai H, Jimenez R, Huang H, Vedell PT, Eckloff BW, Quevedo JF, Pitot HC, Costello BA, Jen J, Wieben ED, Silverstein KAT, Lang JM, Wang L, Dehm SM. Androgen Receptor Variant AR-V9 Is Coexpressed with AR-V7 in Prostate Cancer Metastases and Predicts Abiraterone Resistance. Clin Cancer Res 2017; 23:4704-4715. [PMID: 28473535 PMCID: PMC5644285 DOI: 10.1158/1078-0432.ccr-17-0017] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/13/2017] [Accepted: 04/27/2017] [Indexed: 01/22/2023]
Abstract
Purpose: Androgen receptor (AR) variant AR-V7 is a ligand-independent transcription factor that promotes prostate cancer resistance to AR-targeted therapies. Accordingly, efforts are under way to develop strategies for monitoring and inhibiting AR-V7 in castration-resistant prostate cancer (CRPC). The purpose of this study was to understand whether other AR variants may be coexpressed with AR-V7 and promote resistance to AR-targeted therapies.Experimental Design: We utilized complementary short- and long-read sequencing of intact AR mRNA isoforms to characterize AR expression in CRPC models. Coexpression of AR-V7 and AR-V9 mRNA in CRPC metastases and circulating tumor cells was assessed by RNA-seq and RT-PCR, respectively. Expression of AR-V9 protein in CRPC models was evaluated with polyclonal antisera. Multivariate analysis was performed to test whether AR variant mRNA expression in metastatic tissues was associated with a 12-week progression-free survival endpoint in a prospective clinical trial of 78 CRPC-stage patients initiating therapy with the androgen synthesis inhibitor, abiraterone acetate.Results: AR-V9 was frequently coexpressed with AR-V7. Both AR variant species were found to share a common 3' terminal cryptic exon, which rendered AR-V9 susceptible to experimental manipulations that were previously thought to target AR-V7 uniquely. AR-V9 promoted ligand-independent growth of prostate cancer cells. High AR-V9 mRNA expression in CRPC metastases was predictive of primary resistance to abiraterone acetate (HR = 4.0; 95% confidence interval, 1.31-12.2; P = 0.02).Conclusions: AR-V9 may be an important component of therapeutic resistance in CRPC. Clin Cancer Res; 23(16); 4704-15. ©2017 AACR.
Collapse
Affiliation(s)
- Manish Kohli
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Yeung Ho
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - David W Hillman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Rochester, Minnesota
| | - Jamie L Van Etten
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Christine Henzler
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota
| | - Rendong Yang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota
| | - Jamie M Sperger
- Department of Medicine, Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Yingming Li
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | | | - Ting Hon
- Pacific Biosciences, Menlo Park, California
| | | | - Winston Tan
- Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Rachel E Carlson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Rochester, Minnesota
| | - Liguo Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Hugues Sicotte
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Rochester, Minnesota
| | - Ho Thai
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Rafael Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Peter T Vedell
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Rochester, Minnesota
| | | | - Jorge F Quevedo
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Henry C Pitot
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Brian A Costello
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jin Jen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Medical Genome Facility, Mayo Clinic, Rochester, Minnesota
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Eric D Wieben
- Medical Genome Facility, Mayo Clinic, Rochester, Minnesota
| | | | - Joshua M Lang
- Department of Medicine, Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Scott M Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
- Department of Urology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
18
|
Abstract
Recent studies across multiple tumour types are starting to reveal a recurrent regulatory architecture in which genomic alterations cluster upstream of functional master regulator (MR) proteins, the aberrant activity of which is both necessary and sufficient to maintain tumour cell state. These proteins form small, hyperconnected and autoregulated modules (termed tumour checkpoints) that are increasingly emerging as optimal biomarkers and therapeutic targets. Crucially, as their activity is mostly dysregulated in a post-translational manner, rather than by mutations in their corresponding genes or by differential expression, the identification of MR proteins by conventional methods is challenging. In this Opinion article, we discuss novel methods for the systematic analysis of MR proteins and of the modular regulatory architecture they implement, including their use as a valuable reductionist framework to study the genetic heterogeneity of human disease and to drive key translational applications.
Collapse
Affiliation(s)
- Andrea Califano
- Department of Systems Biology, Columbia University, and the Departments of Biomedical Informatics, Biochemistry and Molecular Biophysics, JP Sulzberger Columbia Genome Center, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Mariano J Alvarez
- DarwinHealth, Inc., 3960 Broadway, Suite 540, New York, New York 10032, USA
| |
Collapse
|
19
|
Ham SL, Joshi R, Thakuri PS, Tavana H. Liquid-based three-dimensional tumor models for cancer research and drug discovery. Exp Biol Med (Maywood) 2016; 241:939-54. [PMID: 27072562 PMCID: PMC4950350 DOI: 10.1177/1535370216643772] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumors are three-dimensional tissues where close contacts between cancer cells, intercellular interactions between cancer and stromal cells, adhesion of cancer cells to the extracellular matrix, and signaling of soluble factors modulate functions of cancer cells and their response to therapeutics. Three-dimensional cultures of cancer cells overcome limitations of traditionally used monolayer cultures and recreate essential characteristics of tumors such as spatial gradients of oxygen, growth factors, and metabolites and presence of necrotic, hypoxic, quiescent, and proliferative cells. As such, three-dimensional tumor models provide a valuable tool for cancer research and oncology drug discovery. Here, we describe different tumor models and primarily focus on a model known as tumor spheroid. We summarize different technologies of spheroid formation, and discuss the use of spheroids to address the influence of stromal fibroblasts and immune cells on cancer cells in tumor microenvironment, study cancer stem cells, and facilitate compound screening in the drug discovery process. We review major techniques for quantification of cellular responses to drugs and discuss challenges ahead to enable broad utility of tumor spheroids in research laboratories, integrate spheroid models into drug development and discovery pipeline, and use primary tumor cells for drug screening studies to realize personalized cancer treatment.
Collapse
Affiliation(s)
- Stephanie L Ham
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Pradip S Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
20
|
Spheroid culture of LuCaP 136 patient-derived xenograft enables versatile preclinical models of prostate cancer. Clin Exp Metastasis 2016; 33:325-37. [PMID: 26873136 DOI: 10.1007/s10585-016-9781-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/10/2016] [Indexed: 12/22/2022]
Abstract
LuCaP serially transplantable patient-derived xenografts (PDXs) are valuable preclinical models of locally advanced or metastatic prostate cancer. Using spheroid culture methodology, we recently established cell lines from several LuCaP PDXs. Here, we characterized in depth the features of xenografts derived from LuCaP 136 spheroid cultures and found faithful retention of the phenotype of the original PDX. In vitro culture enabled luciferase transfection into LuCaP 136 spheroids, facilitating in vivo imaging. We showed that LuCaP 136 spheroids formed intratibial, orthotopic, and subcutaneous tumors when re-introduced into mice. Intratibial tumors responded to castration and were highly osteosclerotic. LuCaP 136 is a realistic in vitro-in vivo preclinical model of a subtype of bone metastatic prostate cancer.
Collapse
|
21
|
Borges GT, Vêncio EF, Quek SI, Chen A, Salvanha DM, Vêncio RZN, Nguyen HM, Vessella RL, Cavanaugh C, Ware CB, Troisch P, Liu AY. Conversion of Prostate Adenocarcinoma to Small Cell Carcinoma-Like by Reprogramming. J Cell Physiol 2016; 231:2040-7. [PMID: 26773436 DOI: 10.1002/jcp.25313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/14/2022]
Abstract
The lineage relationship between prostate adenocarcinoma and small cell carcinoma was studied by using the LuCaP family of xenografts established from primary neoplasm to metastasis. Expression of four stem cell transcription factor (TF) genes, LIN28A, NANOG, POU5F1, SOX2, were analyzed in the LuCaP lines. These genes, when force expressed in differentiated cells, can reprogram the recipients into stem-like induced pluripotent stem (iPS) cells. Most LuCaP lines expressed POU5F1, while LuCaP 145.1, representative of small cell carcinoma, expressed all four. Through transcriptome database query, many small cell carcinoma genes were also found in stem cells. To test the hypothesis that prostate cancer progression from "differentiated" adenocarcinoma to "undifferentiated" small cell carcinoma could involve re-expression of stem cell genes, the four TF genes were transduced via lentiviral vectors into five adenocarcinoma LuCaP lines-70CR, 73CR, 86.2, 92, 105CR-as done in iPS cell reprogramming. The resultant cells from these five transductions displayed a morphology of small size and dark appearing unlike the parentals. Transcriptome analysis of LuCaP 70CR* ("*" to denote transfected progeny) revealed a unique gene expression close to that of LuCaP 145.1. In a prostate principal components analysis space based on cell-type transcriptomes, the different LuCaP transcriptome datapoints were aligned to suggest a possible ordered sequence of expression changes from the differentiated luminal-like adenocarcinoma cell types to the less differentiated, more stem-like small cell carcinoma types, and LuCaP 70CR*. Prostate cancer progression can thus be molecularly characterized by loss of differentiation with re-expression of stem cell genes. J. Cell. Physiol. 231: 2040-2047, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gisely T Borges
- Department of Urology, University of Washington, Seattle, Washington.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Pharmacy School, Federal University of Goiás, Goiânia, Brazil
| | - Eneida F Vêncio
- Department of Urology, University of Washington, Seattle, Washington.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Oral Pathology, Dental School, Federal University of Goiás, Goiânia, Brazil
| | - Sue-Ing Quek
- Department of Urology, University of Washington, Seattle, Washington.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Adeline Chen
- Department of Urology, University of Washington, Seattle, Washington.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Diego M Salvanha
- Department of Computing and Mathematics, FFCLRP, University of São Paulo, São Paulo, Brazil
| | - Ricardo Z N Vêncio
- Department of Computing and Mathematics, FFCLRP, University of São Paulo, São Paulo, Brazil
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, Washington
| | - Robert L Vessella
- Department of Urology, University of Washington, Seattle, Washington.,Puget Sound VA Medical Center, Seattle, Washington
| | - Christopher Cavanaugh
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Carol B Ware
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Comparative Medicine, University of Washington, Seattle, Washington
| | | | - Alvin Y Liu
- Department of Urology, University of Washington, Seattle, Washington.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
22
|
Weiswald LB, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. Neoplasia 2015; 17:1-15. [PMID: 25622895 PMCID: PMC4309685 DOI: 10.1016/j.neo.2014.12.004] [Citation(s) in RCA: 779] [Impact Index Per Article: 86.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 11/29/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional (3D) in vitro models have been used in cancer research as an intermediate model between in vitro cancer cell line cultures and in vivo tumor. Spherical cancer models represent major 3D in vitro models that have been described over the past 4 decades. These models have gained popularity in cancer stem cell research using tumorospheres. Thus, it is crucial to define and clarify the different spherical cancer models thus far described. Here, we focus on in vitro multicellular spheres used in cancer research. All these spherelike structures are characterized by their well-rounded shape, the presence of cancer cells, and their capacity to be maintained as free-floating cultures. We propose a rational classification of the four most commonly used spherical cancer models in cancer research based on culture methods for obtaining them and on subsequent differences in sphere biology: the multicellular tumor spheroid model, first described in the early 70s and obtained by culture of cancer cell lines under nonadherent conditions; tumorospheres, a model of cancer stem cell expansion established in a serum-free medium supplemented with growth factors; tissue-derived tumor spheres and organotypic multicellular spheroids, obtained by tumor tissue mechanical dissociation and cutting. In addition, we describe their applications to and interest in cancer research; in particular, we describe their contribution to chemoresistance, radioresistance, tumorigenicity, and invasion and migration studies. Although these models share a common 3D conformation, each displays its own intrinsic properties. Therefore, the most relevant spherical cancer model must be carefully selected, as a function of the study aim and cancer type.
Collapse
Affiliation(s)
- Louis-Bastien Weiswald
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Laboratoire d'Oncobiologie, Hôpital René Huguenin, Institut Curie, St Cloud, France; Université Paris Descartes, Faculté de Pharmacie de Paris, Sorbonne Paris Cité, Paris, France.
| | - Dominique Bellet
- Laboratoire d'Oncobiologie, Hôpital René Huguenin, Institut Curie, St Cloud, France; Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, UMR 8151 CNRS-U1022 Inserm, Sorbonne Paris Cité, Paris, France
| | - Virginie Dangles-Marie
- Université Paris Descartes, Faculté de Pharmacie de Paris, Sorbonne Paris Cité, Paris, France; Département de Recherche Translationnelle, Research Center, Institut Curie, Paris, France.
| |
Collapse
|
23
|
Lawrence MG, Pook DW, Wang H, Porter LH, Frydenberg M, Kourambas J, Appu S, Poole C, Beardsley EK, Ryan A, Norden S, Papargiris MM, Risbridger GP, Taylor RA. Establishment of primary patient-derived xenografts of palliative TURP specimens to study castrate-resistant prostate cancer. Prostate 2015; 75:1475-83. [PMID: 26177841 DOI: 10.1002/pros.23039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/26/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fresh patient specimens of castrate-resistant prostate cancer (CRPC) are invaluable for studying tumor heterogeneity and responses to current treatments. They can be used for primary patient-derived xenografts (PDXs) or serially transplantable PDXs, but only a small proportion of samples grow successfully. To improve the efficiency and quality of PDXs, we investigated the factors that determine the initial engraftment of patient tissues derived from TURP specimens. METHODS Fresh tissue was collected from castrate patients who required a TURP for urinary symptoms. Tissue was grafted under the renal capsule of immune-compromised mice for up to 14 weeks. The abundance of cancer in ungrafted and grafted specimens was compared using histopathology. Mice were castrated or implanted with testosterone pellets to determine the androgen-responsiveness of CRPC PDXs from TURP tissue. RESULTS Primary PDXs were successfully established from 7 of 10 patients that underwent grafting. Of the 112 grafts generated from these 10 patients, 21% contained cancer at harvest. Grafts were most successful when the original patient specimens contained high amounts of viable cancer, defined as samples with (i) at least 50% cancer cells, (ii) no physical damage, and (iii) detectable Ki67 expression. PDX grafts survived in castrated hosts and proliferated in response to testosterone, confirming that they were castrate resistant but androgen-responsive. CONCLUSIONS Primary PDXs of CRPC can be established from TURP specimens with modest success. The take rate can be increased if the original tissues contain sufficient numbers of actively proliferating cancer cells. Selecting specimens with abundant viable cancer will maximize the rate of engraftment and increase the efficiency of establishing PDXs that can be serially transplanted.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - David W Pook
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Department of Oncology, Monash Cancer Centre, Melbourne, Victoria, Australia
| | - Hong Wang
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Mark Frydenberg
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Department of Urology, Monash Medical Centre, Clayton, Victoria, Australia
- Department of Surgery, Monash University, Clayton, Victoria, Australia
| | - John Kourambas
- Department of Urology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Sree Appu
- Department of Urology, Monash Medical Centre, Clayton, Victoria, Australia
- Department of Surgery, Monash University, Clayton, Victoria, Australia
| | - Christine Poole
- Department of Urology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Emma K Beardsley
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Andrew Ryan
- TissuPath Pathology, Melbourne, Mount Waverley, Victoria, Australia
| | - Sam Norden
- TissuPath Pathology, Melbourne, Mount Waverley, Victoria, Australia
| | - Melissa M Papargiris
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Renea A Taylor
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Borges GT, Vêncio EF, Vêncio RZN, Vessella RL, Ware CB, Liu AY. Reprogramming of prostate cancer cells--technical challenges. Curr Urol Rep 2015; 16:468. [PMID: 25404182 DOI: 10.1007/s11934-014-0468-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prostate cancer progression is characterized by tumor dedifferentiation. Cancer cells of less differentiated tumors have a gene expression/transcriptome more similar to that of stem cells. In dedifferentiation, cancer cells may follow a specific program of gene expression changes to a stem-like state. In order to treat cancer effectively, the stem-like cancer cells and cancer differentiation pathway need to be identified and studied. Due to the very low abundance of stem-like cancer cells, their isolation from fresh human tumors is technically challenging. Induced pluripotent stem cell technology can reprogram differentiated cells into stem-like, and this may be a tool to generate sufficient stem-like cancer cells.
Collapse
Affiliation(s)
- Gisely T Borges
- School of Pharmacology, Federal University of Goiás, Goiânia, Brazil,
| | | | | | | | | | | |
Collapse
|
25
|
Ruppender N, Larson S, Lakely B, Kollath L, Brown L, Coleman I, Coleman R, Nguyen H, Nelson PS, Corey E, Snyder LA, Vessella RL, Morrissey C, Lam HM. Cellular Adhesion Promotes Prostate Cancer Cells Escape from Dormancy. PLoS One 2015; 10:e0130565. [PMID: 26090669 PMCID: PMC4475050 DOI: 10.1371/journal.pone.0130565] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/21/2015] [Indexed: 12/20/2022] Open
Abstract
Dissemination of prostate cancer (PCa) cells to the bone marrow is an early event in the disease process. In some patients, disseminated tumor cells (DTC) proliferate to form active metastases after a prolonged period of undetectable disease known as tumor dormancy. Identifying mechanisms of PCa dormancy and reactivation remain a challenge partly due to the lack of in vitro models. Here, we characterized in vitro PCa dormancy-reactivation by inducing cells from three patient-derived xenograft (PDX) lines to proliferate through tumor cell contact with each other and with bone marrow stroma. Proliferating PCa cells demonstrated tumor cell-cell contact and integrin clustering by immunofluorescence. Global gene expression analyses on proliferating cells cultured on bone marrow stroma revealed a downregulation of TGFB2 in all of the three proliferating PCa PDX lines when compared to their non-proliferating counterparts. Furthermore, constitutive activation of myosin light chain kinase (MLCK), a downstream effector of integrin-beta1 and TGF-beta2, in non-proliferating cells promoted cell proliferation. This cell proliferation was associated with an upregulation of CDK6 and a downregulation of E2F4. Taken together, our data provide the first clinically relevant in vitro model to support cellular adhesion and downregulation of TGFB2 as a potential mechanism by which PCa cells may escape from dormancy. Targeting the TGF-beta2-associated mechanism could provide novel opportunities to prevent lethal PCa metastasis.
Collapse
Affiliation(s)
- Nazanin Ruppender
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Sandy Larson
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Bryce Lakely
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Lori Kollath
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Lisha Brown
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Ilsa Coleman
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Roger Coleman
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Holly Nguyen
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Linda A. Snyder
- Janssen Research and Development, LLC, Spring House, Pennsylvania, United States of America
| | - Robert L. Vessella
- Department of Urology, University of Washington, Seattle, Washington, United States of America
- Department of Veterans Affairs Medical Center, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
26
|
Abstract
INTRODUCTION The mouse is an important, though imperfect, organism with which to model human disease and to discover and test novel drugs in a preclinical setting. Many experimental strategies have been used to discover new biological and molecular targets in the mouse, with the hopes of translating these discoveries into novel drugs to treat prostate cancer in humans. Modeling prostate cancer in the mouse, however, has been challenging, and often drugs that work in mice have failed in human trials. AREAS COVERED The authors discuss the similarities and differences between mice and men; the types of mouse models that exist to model prostate cancer; practical questions one must ask when using a mouse as a model; and potential reasons that drugs do not often translate to humans. They also discuss the current value in using mouse models for drug discovery to treat prostate cancer and what needs are still unmet in field. EXPERT OPINION With proper planning and following practical guidelines by the researcher, the mouse is a powerful experimental tool. The field lacks genetically engineered metastatic models, and xenograft models do not allow for the study of the immune system during the metastatic process. There remain several important limitations to discovering and testing novel drugs in mice for eventual human use, but these can often be overcome. Overall, mouse modeling is an essential part of prostate cancer research and drug discovery. Emerging technologies and better and ever-increasing forms of communication are moving the field in a hopeful direction.
Collapse
Affiliation(s)
- Kenneth C Valkenburg
- The Johns Hopkins University, The James Buchanan Brady Urological Institute, Department of Urology , 600 North Wolfe Street, Baltimore, MD 21287 , USA
| | | |
Collapse
|
27
|
Saar M, Zhao H, Nolley R, Young SR, Coleman I, Nelson PS, Vessella RL, Peehl DM. Spheroid culture of LuCaP 147 as an authentic preclinical model of prostate cancer subtype with SPOP mutation and hypermutator phenotype. Cancer Lett 2014; 351:272-80. [PMID: 24998678 DOI: 10.1016/j.canlet.2014.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/16/2014] [Accepted: 06/24/2014] [Indexed: 11/24/2022]
Abstract
LuCaP serially transplantable xenografts are valuable preclinical models of locally advanced or metastatic prostate cancer. For the first time, we recently succeeded in establishing and serially passaging spheroid cultures of several LuCaP xenografts. Here, we characterized in depth the molecular and cellular phenotype of LuCaP 147 cultures and found faithful retention of the characteristics of the original xenograft, including immunophenotype, genetic fidelity, gene expression profile and responsiveness to androgen. Furthermore, we demonstrated capabilities for high-throughput drug screening and that anti-cancer agents induced cell cycle arrest and apoptosis in spheroid cultures. Finally, we showed that cells formed tumors when re-introduced into mice, providing an authentic in vitro-in vivo preclinical model of a subtype of prostate cancer with a hypermutator phenotype and an SPOP mutation.
Collapse
Affiliation(s)
- Matthias Saar
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Urology and Pediatric Urology, University of Saarland, Homburg, Saar, Germany
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rosalie Nolley
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sarah R Young
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robert L Vessella
- Department of Urology, University of Washington, Puget Sound VA Health Care System, Seattle, WA 98195, USA
| | - Donna M Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|