1
|
Chiu CL, Zhang D, Zhao H, Wei Y, Polasko AL, Thomsen MT, Yang V, Yang KK, Hauck S, Peterson EE, Wen RM, Qiu Z, Corey E, Miao YR, Rankin EB, Peehl DM, Huang J, Giaccia AJ, Brooks JD. Targeting AXL Inhibits the Growth and Metastasis of Prostate Cancer in Bone. Clin Cancer Res 2025; 31:1346-1358. [PMID: 39879384 PMCID: PMC11961319 DOI: 10.1158/1078-0432.ccr-24-3028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in prostate cancer and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. EXPERIMENTAL DESIGN Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by IHC staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on tumor growth and lung metastases in prostate cancer patient-derived xenograft models that were implanted intratibially. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum prostate-specific antigen or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lungs was determined using qPCR. Transcriptomic profiling, Western blotting, and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. RESULTS High AXL phosphorylation in human prostate cancer bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lungs through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, and ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. CONCLUSIONS Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal metastatic prostate cancer.
Collapse
Affiliation(s)
- Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Wei
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Mikkel Thy Thomsen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Vanessa Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasie Kexin Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Hauck
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Eric E. Peterson
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ru M. Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhengyuan Qiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Oncology, University of Oxford, Oxford, UK
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
2
|
Corres-Mendizabal J, Zacchi F, Martín-Martín N, Mateo J, Carracedo A. Metastatic hormone-naïve prostate cancer: a distinct biological entity. Trends Cancer 2024; 10:825-841. [PMID: 39048488 PMCID: PMC11397905 DOI: 10.1016/j.trecan.2024.06.005] [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: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Metastatic hormone-naïve prostate cancer (mHNPC) is often the initial form of presentation for metastatic prostate cancer and encompasses a heterogeneous patient population with high inter-patient heterogeneity in prognosis and response to therapy. A more precise treatment of mHNPC, guided by evidence-based biomarkers, remains an unmet medical need. In addition, the limited number of representative laboratory models of mHNPC hampers the translation of basic research into clinical applications. We provide a comprehensive overview of the clinical and biological features that characterize mHNPC, highlight molecular data that could explain the unique prognostic characteristics of mHNPC, and identify key open questions.
Collapse
Affiliation(s)
- Jon Corres-Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Francesca Zacchi
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy; Vall Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital Campus, Barcelona, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; Translational Prostate Cancer Research Laboratory, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Joaquin Mateo
- Vall Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital Campus, Barcelona, Spain.
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; Translational Prostate Cancer Research Laboratory, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
3
|
Yin J, Daryanani A, Lu F, Ku AT, Bright JR, Alilin ANS, Bowman J, Lake R, Li C, Truong TM, Twohig JD, Mostaghel EA, Ishikawa M, Simpson M, Trostel SY, Corey E, Sowalsky AG, Kelly K. Reproducible preclinical models of androgen receptor driven human prostate cancer bone metastasis. Prostate 2024; 84:1033-1046. [PMID: 38708958 PMCID: PMC11216894 DOI: 10.1002/pros.24718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Preclinical models recapitulating the metastatic phenotypes are essential for developing the next-generation therapies for metastatic prostate cancer (mPC). We aimed to establish a cohort of clinically relevant mPC models, particularly androgen receptor positive (AR+) bone metastasis models, from LuCaP patient-derived xenografts (PDX) that reflect the heterogeneity and complexity of mPC. METHODS PDX tumors were dissociated into single cells, modified to express luciferase, and were inoculated into NSG mice via intracardiac injection. The progression of metastases was monitored by bioluminescent imaging. Histological phenotypes of metastases were characterized by immunohistochemistry and immunofluorescence staining. Castration responses were further investigated in two AR-positive models. RESULTS Our PDX-derived metastasis (PDM) model collection comprises three AR+ adenocarcinomas (ARPC) and one AR- neuroendocrine carcinoma (NEPC). All ARPC models developed bone metastases with either an osteoblastic, osteolytic, or mixed phenotype, while the NEPC model mainly developed brain metastasis. Different mechanisms of castration resistance were observed in two AR+ PDM models with distinct genotypes, such as combined loss of TP53 and RB1 in one model and expression of AR splice variant 7 (AR-V7) expression in another model. Intriguingly, the castration-resistant tumors displayed inter- and intra-tumor as well as organ-specific heterogeneity in lineage specification. CONCLUSION Genetically diverse PDM models provide a clinically relevant system for biomarker identification and personalized medicine in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- JuanJuan Yin
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Asha Daryanani
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Fan Lu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Anson T. Ku
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - John R. Bright
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Aian Neil S. Alilin
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Joel Bowman
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Ross Lake
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Chennan Li
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Tri M. Truong
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Joseph D. Twohig
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Masaki Ishikawa
- Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mark Simpson
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Shana Y. Trostel
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Adam G. Sowalsky
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
4
|
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
|
5
|
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 PMCID: PMC12016311 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] [Grants] [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
|
6
|
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
|
7
|
Gnatowski P, Piłat E, Kucińska-Lipka J, Saeb MR, Hamblin MR, Mozafari M. Recent advances in 3D bioprinted tumor models for personalized medicine. Transl Oncol 2023; 37:101750. [PMID: 37572498 PMCID: PMC10440569 DOI: 10.1016/j.tranon.2023.101750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/14/2023] Open
Abstract
Cancerous tumors are among the most fatal diseases worldwide, claiming nearly 10 million lives in 2020. Due to their complex and dynamic nature, modeling tumors accurately is a challenging task. Current models suffer from inadequate translation between in vitro and in vivo results, primarily due to the isotropic nature of tumors and their microenvironment's relationship. To address these limitations, hydrogel-based 3D bioprinting is emerging as a promising approach to mimic cancer development and behavior. It provides precise control over individual elements' size and distribution within the cancer microenvironment and enables the use of patient-derived tumor cells, rather than commercial lines. Consequently, hydrogel bioprinting is expected to become a state-of-the-art technique for cancer research. This manuscript presents an overview of cancer statistics, current modeling methods, and their limitations. Additionally, we highlight the significance of bioprinting, its applications in cancer modeling, and the importance of hydrogel selection. We further explore the current state of creating models for the five deadliest cancers using 3D bioprinting. Finally, we discuss current trends and future perspectives on the clinical use of cancer modeling using hydrogel bioprinting.
Collapse
Affiliation(s)
- Przemysław Gnatowski
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Edyta Piłat
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Justyna Kucińska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
8
|
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: 24] [Impact Index Per Article: 8.0] [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
|
9
|
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: 11] [Impact Index Per Article: 3.7] [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
|
10
|
Gündel B, Liu X, Löhr M, Heuchel R. Pancreatic Ductal Adenocarcinoma: Preclinical in vitro and ex vivo Models. Front Cell Dev Biol 2021; 9:741162. [PMID: 34746135 PMCID: PMC8569794 DOI: 10.3389/fcell.2021.741162] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most overlooked cancers despite its dismal median survival time of 6 months. The biggest challenges in improving patient survival are late diagnosis due to lack of diagnostic markers, and limited treatment options due to almost complete therapy resistance. The past decades of research identified the dense stroma and the complex interplay/crosstalk between the cancer- and the different stromal cells as the main culprits for the slow progress in improving patient outcome. For better ex vivo simulation of this complex tumor microenvironment the models used in PDAC research likewise need to become more diverse. Depending on the focus of the investigation, several in vitro and in vivo models for PDAC have been established in the past years. Particularly, 3D cell culture such as spheroids and organoids have become more frequently used. This review aims to examine current PDAC in vitro models, their inherent limitations, and their successful implementations in research.
Collapse
Affiliation(s)
- Beate Gündel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Xinyuan Liu
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Matthias Löhr
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
- Department of Upper GI, C1:77, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Rainer Heuchel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
11
|
Risbridger GP, Lawrence MG, Taylor RA. PDX: Moving Beyond Drug Screening to Versatile Models for Research Discovery. J Endocr Soc 2020; 4:bvaa132. [PMID: 33094211 PMCID: PMC7566391 DOI: 10.1210/jendso/bvaa132] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 01/08/2023] Open
Abstract
Patient-derived xenografts (PDXs) are tools of the trade for many researchers from all disciplines and medical specialties. Most endocrinologists, and especially those working in oncology, commonly use PDXs for preclinical drug testing and development, and over the last decade large collections of PDXs have emerged across all tumor streams. In this review, we examine how the field has evolved to include PDXs as versatile resources for research discoveries, providing evidence for guidelines and changes in clinical practice.
Collapse
Affiliation(s)
- Gail P Risbridger
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Physiology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
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: 24] [Impact Index Per Article: 4.8] [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
|
13
|
Huang Y, Lu Y, Vadlamudi M, Zhao S, Felmlee M, Rahimian R, Guo X. Intrapulmonary inoculation of multicellular spheroids to construct an orthotopic lung cancer xenograft model that mimics four clinical stages of non-small cell lung cancer. J Pharmacol Toxicol Methods 2020; 104:106885. [PMID: 32531198 DOI: 10.1016/j.vascn.2020.106885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Lung cancer leads in mortality among all types of cancer in US and Non-small cell lung cancer (NSCLC) is the major type of lung cancer. Mice models of lung cancer based on subcutaneous or orthotopic inoculation of cancer cell suspension do not adequately mimic the progression of lung cancer in clinic. METHODS A549-iRFP cells (human NSCLC adenocarcinoma) were cultured to form multicellular spheroids (MCS), which were then inoculated intrapulmonarily into male athymic nude mice. The xenograft cancer development was monitored by in vivo fluorescent imaging and validated by open-chest anatomy, ex vivo fluorescent imaging, and histological studies. RESULTS The newly developed orthotopic xenograft model of lung cancer simulated all four clinical stages of NSCLC progression over one month: Stage 1) localized tumor at the inoculation site, Stage 2) multiple tumor nodules or larger tumor nodule on the same side of the lung, Stage 3) cancer growth on heart surface, and Stage 4) metastatic cancer on both sides of the lung. The model yielded high rates of postoperative survival (100%) and parenchymal tumor establishment (88.9%). The roughness of the inoculated MCS associated negatively with the time needed to develop metastatic cancer (p = .0299). DISCUSSION This new orthotopic xenograft model of NSCLC would facilitate the development of medications to treat lung cancer.
Collapse
Affiliation(s)
- Yingbo Huang
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Yifan Lu
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Mallika Vadlamudi
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Shen Zhao
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Melanie Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Roshanak Rahimian
- Department of Physiology and Pharmacology, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| | - Xin Guo
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA.
| |
Collapse
|
14
|
Valta M, Ylä-Pelto J, Lan Y, Kähkönen T, Taimen P, Boström PJ, Ettala O, Khan S, Paulin N, Elo LL, Koskinen PJ, Härkönen P, Tuomela J. Critical evaluation of the subcutaneous engraftments of hormone naïve primary prostate cancer. Transl Androl Urol 2020; 9:1120-1134. [PMID: 32676396 PMCID: PMC7354344 DOI: 10.21037/tau.2020.03.38] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Patient-derived xenografts (PDXs) are considered to better recapitulate the histopathological and molecular heterogeneity of human cancer than other preclinical models. Despite technological advances, PDX models from hormone naïve primary prostate cancer are scarce. We performed a detailed analysis of PDX methodology using a robust subcutaneous model and fresh tissues from patients with primary hormone naïve prostate cancer. Methods Clinical prostate tumor specimens (n=26, Gleason score 6-10) were collected from robotic-assisted laparoscopic radical prostatectomies at Turku University Hospital (Turku, Finland), cut into pieces, and implanted subcutaneously into 84 immunodeficient mice. Engraftments and the adjacent material from prostatic surgical specimens were compared using histology, immunohistochemistry and DNA sequencing. Results The probability of a successful engraftment correlated with the presence of carcinoma in the implanted tissue. Tumor take rate was 41%. Surprisingly, mouse hormone supplementation inhibited tumor take rate, whereas the degree of mouse immunodeficiency did not have an effect. Histologically, the engrafted tumors closely mimicked their parental tumors, and the Gleason grades and copy number variants of the engraftments were similar to those of their primary tumors. Expression levels of androgen receptor, prostate-specific antigen, and keratins were retained in engraftments, and a detailed genomic analysis revealed high fidelity of the engraftments with their corresponding primary tumors. However, in the second or third passage of tumors, the carcinoma areas were almost completely replaced by benign tissue with frequent degenerative or metaplastic changes. Conclusions Subcutaneous primary prostate engraftments preserve the phenotypic and genotypic landscape. Thus, they serve a potential model for personalized medicine and preclinical research but their use may be limited to the first passage.
Collapse
Affiliation(s)
- Maija Valta
- Institute of Biomedicine, University of Turku, Turku, Finland.,Division of Medicine, Turku City Hospital, Turku, Finland
| | - Jani Ylä-Pelto
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Biology, University of Turku, Turku, Finland
| | - Yu Lan
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tiina Kähkönen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pekka Taimen
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| | - Peter J Boström
- Department of Urology, Turku University Hospital and University of Turku, Turku, Finland
| | - Otto Ettala
- Department of Urology, Turku University Hospital and University of Turku, Turku, Finland
| | - Sofia Khan
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Niklas Paulin
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Laura L Elo
- Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Pirkko Härkönen
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN WEST Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Johanna Tuomela
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN WEST Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
15
|
Risbridger GP, Toivanen R, Taylor RA. Preclinical Models of Prostate Cancer: Patient-Derived Xenografts, Organoids, and Other Explant Models. Cold Spring Harb Perspect Med 2018; 8:a030536. [PMID: 29311126 PMCID: PMC6071547 DOI: 10.1101/cshperspect.a030536] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prostate cancer remains a lethal disease. Preclinical cancer models that accurately represent the tumors of the patients they are intended to help are necessary to test potential therapeutic approaches and to better translate research discoveries. However, research in the prostate cancer field is hampered by the limited number of human cell lines and xenograft models, most of which do not recapitulate the human disease seen in the clinic today. This work reviews the recent advances in human patient-derived xenograft, organoid, and other explant models to address this need. In contrast to other tumor streams, the prostate cancer field is challenged by this approach, yet despite the limitations, patient-derived models remain an integral component of the preclinical testing pathway leading to better treatments for men with prostate cancer.
Collapse
Affiliation(s)
- Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Melbourne, Victoria 3168, Australia
- Cancer Discovery Program, Biomedicine Discovery Institute; Prostate Cancer Research Group, Department of Anatomy and Developmental Biology; and Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia
- Prostate Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Roxanne Toivanen
- Prostate Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria 3000, Australia
- Departments of Medicine, Genetics & Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Renea A Taylor
- Cancer Discovery Program, Biomedicine Discovery Institute; Prostate Cancer Research Group, Department of Anatomy and Developmental Biology; and Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia
- Prostate Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria 3000, Australia
| |
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: 18] [Impact Index Per Article: 2.6] [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
|
|
18
|
Rodenhizer D, Dean T, D'Arcangelo E, McGuigan AP. The Current Landscape of 3D In Vitro Tumor Models: What Cancer Hallmarks Are Accessible for Drug Discovery? Adv Healthc Mater 2018; 7:e1701174. [PMID: 29350495 DOI: 10.1002/adhm.201701174] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/16/2017] [Indexed: 12/11/2022]
Abstract
Cancer prognosis remains a lottery dependent on cancer type, disease stage at diagnosis, and personal genetics. While investment in research is at an all-time high, new drugs are more likely to fail in clinical trials today than in the 1970s. In this review, a summary of current survival statistics in North America is provided, followed by an overview of the modern drug discovery process, classes of models used throughout different stages, and challenges associated with drug development efficiency are highlighted. Then, an overview of the cancer hallmarks that drive clinical progression is provided, and the range of available clinical therapies within the context of these hallmarks is categorized. Specifically, it is found that historically, the development of therapies is limited to a subset of possible targets. This provides evidence for the opportunities offered by novel disease-relevant in vitro models that enable identification of novel targets that facilitate interactions between the tumor cells and their surrounding microenvironment. Next, an overview of the models currently reported in literature is provided, and the cancer biology they have been used to explore is highlighted. Finally, four priority areas are suggested for the field to accelerate adoption of in vitro tumour models for cancer drug discovery.
Collapse
Affiliation(s)
- Darren Rodenhizer
- Department of Chemical Engineering and Applied ChemistryUniversity of Toronto 200 College Street Toronto M5S 3E5 Canada
| | - Teresa Dean
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto 200 College Street Toronto M5S 3E5 Canada
| | - Elisa D'Arcangelo
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto 200 College Street Toronto M5S 3E5 Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry & Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto 200 College Street Toronto M5S 3E5 Canada
| |
Collapse
|
19
|
Fong ELS, Harrington DA, Farach-Carson MC, Yu H. Heralding a new paradigm in 3D tumor modeling. Biomaterials 2016; 108:197-213. [PMID: 27639438 DOI: 10.1016/j.biomaterials.2016.08.052] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022]
Abstract
Numerous studies to date have contributed to a paradigm shift in modeling cancer, moving from the traditional two-dimensional culture system to three-dimensional (3D) culture systems for cancer cell culture. This led to the inception of tumor engineering, which has undergone rapid advances over the years. In line with the recognition that tumors are not merely masses of proliferating cancer cells but rather, highly complex tissues consisting of a dynamic extracellular matrix together with stromal, immune and endothelial cells, significant efforts have been made to better recapitulate the tumor microenvironment in 3D. These approaches include the development of engineered matrices and co-cultures to replicate the complexity of tumor-stroma interactions in vitro. However, the tumor engineering and cancer biology fields have traditionally relied heavily on the use of cancer cell lines as a cell source in tumor modeling. While cancer cell lines have contributed to a wealth of knowledge in cancer biology, the use of this cell source is increasingly perceived as a major contributing factor to the dismal failure rate of oncology drugs in drug development. Backing this notion is the increasing evidence that tumors possess intrinsic heterogeneity, which predominantly homogeneous cancer cell lines poorly reflect. Tumor heterogeneity contributes to therapeutic resistance in patients. To overcome this limitation, cancer cell lines are beginning to be replaced by primary tumor cell sources, in the form of patient-derived xenografts and organoids cultures. Moving forward, we propose that further advances in tumor engineering would require that tumor heterogeneity (tumor variants) be taken into consideration together with tumor complexity (tumor-stroma interactions). In this review, we provide a comprehensive overview of what has been achieved in recapitulating tumor complexity, and discuss the importance of incorporating tumor heterogeneity into 3D in vitro tumor models. This work carves out the roadmap for 3D tumor engineering and highlights some of the challenges that need to be addressed as we move forward into the next chapter.
Collapse
Affiliation(s)
- Eliza L S Fong
- Department of Physiology, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore.
| | | | | | - Hanry Yu
- Department of Physiology, National University of Singapore, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research, Singapore; Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Risbridger GP, Taylor RA. Patient-Derived Prostate Cancer: from Basic Science to the Clinic. Discov Oncol 2016; 7:236-40. [PMID: 27177552 DOI: 10.1007/s12672-016-0266-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/26/2016] [Indexed: 12/22/2022] Open
Abstract
Systems that model cancer form the backbone of research discovery, and their accuracy and validity are a key determinant to ensure successful translation. In many tumour types, patient-derived specimens are an important model of choice for pre-clinical drug development. In this review, we consider why this has been such a challenge for prostate cancer, resulting in relatively few patient-derived xenografts (PDXs) of prostatic tumours compared to breast cancers, for example. Nevertheless, with only a few patient specimens and PDXs, we exemplify in three vignettes how important new clinical insights were obtained resulting in benefit for future men with prostate cancer.
Collapse
Affiliation(s)
- Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium and Cancer Program, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Melbourne, VIC, 3800, Australia.
| | - Renea A Taylor
- Monash Partners Comprehensive Cancer Consortium and Cancer Program, Biomedicine Discovery Institute, Department of Physiology, Monash University, Wellington Road, Melbourne, VIC, 3800, Australia
| |
Collapse
|