1
|
Reuvers TGA, Grandia V, Brandt RMC, Arab M, Maas SLN, Bos EM, Nonnekens J. Investigating the Radiobiological Response to Peptide Receptor Radionuclide Therapy Using Patient-Derived Meningioma Spheroids. Cancers (Basel) 2024; 16:2515. [PMID: 39061156 PMCID: PMC11275064 DOI: 10.3390/cancers16142515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTA-TATE has recently been evaluated for the treatment of meningioma patients. However, current knowledge of the underlying radiation biology is limited, in part due to the lack of appropriate in vitro models. Here, we demonstrate proof-of-concept of a meningioma patient-derived 3D culture model to assess the short-term response to radiation therapies such as PRRT and external beam radiotherapy (EBRT). We established short-term cultures (1 week) for 16 meningiomas with high efficiency and yield. In general, meningioma spheroids retained characteristics of the parental tumor during the initial days of culturing. For a subset of tumors, clear changes towards a more aggressive phenotype were visible over time, indicating that the culture method induced dedifferentiation of meningioma cells. To assess PRRT efficacy, we demonstrated specific uptake of 177Lu-DOTA-TATE via somatostatin receptor subtype 2 (SSTR2), which was highly overexpressed in the majority of tumor samples. PRRT induced DNA damage which was detectable for an extended timeframe as compared to EBRT. Interestingly, levels of DNA damage in spheroids after PRRT correlated with SSTR2-expression levels of parental tumors. Our patient-derived meningioma culture model can be used to assess the short-term response to PRRT and EBRT in radiobiological studies. Further improvement of this model should pave the way towards the development of a relevant culture model for assessment of the long-term response to radiation and, potentially, individual patient responses to PRRT and EBRT.
Collapse
Affiliation(s)
- Thom G A Reuvers
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Vivian Grandia
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Majd Arab
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sybren L N Maas
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Eelke M Bos
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
2
|
Martinez-Ruiz L, López-Rodríguez A, Florido J, Rodríguez-Santana C, Rodríguez Ferrer JM, Acuña-Castroviejo D, Escames G. Patient-derived tumor models in cancer research: Evaluation of the oncostatic effects of melatonin. Biomed Pharmacother 2023; 167:115581. [PMID: 37748411 DOI: 10.1016/j.biopha.2023.115581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
The development of new anticancer therapies tends to be very slow. Although their impact on potential candidates is confirmed in preclinical studies, ∼95 % of these new therapies are not approved when tested in clinical trials. One of the main reasons for this is the lack of accurate preclinical models. In this context, there are different patient-derived models, which have emerged as a powerful oncological tool: patient-derived xenografts (PDXs), patient-derived organoids (PDOs), and patient-derived cells (PDCs). Although all these models are widely applied, PDXs, which are created by engraftment of patient tumor tissues into mice, is considered more reliable. In fundamental research, the PDX model is used to evaluate drug-sensitive markers and, in clinical practice, to select a personalized therapeutic strategy. Melatonin is of particular importance in the development of innovative cancer treatments due to its oncostatic impact and lack of adverse effects. However, the literature regarding the oncostatic effect of melatonin in patient-derived tumor models is scant. This review aims to describe the important role of patient-derived models in the development of anticancer treatments, focusing, in particular, on PDX models, as well as their use in cancer research. This review also summarizes the existing literature on the anti-tumoral effect of melatonin in patient-derived models in order to propose future anti-neoplastic clinical applications.
Collapse
Affiliation(s)
- Laura Martinez-Ruiz
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Alba López-Rodríguez
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Javier Florido
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Cesar Rodríguez-Santana
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - José M Rodríguez Ferrer
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Germaine Escames
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain.
| |
Collapse
|
3
|
Mui M, Clark M, Vu TMSH, Clemons N, Hollande F, Roth S, Ramsay R, Michael M, Heriot AG, Kong JCH. Use of patient-derived explants as a preclinical model for precision medicine in colorectal cancer: A scoping review. Langenbecks Arch Surg 2023; 408:392. [PMID: 37816905 PMCID: PMC10564805 DOI: 10.1007/s00423-023-03133-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023]
Abstract
PURPOSE Whilst the treatment paradigm for colorectal cancer has evolved significantly over time, there is still a lack of reliable biomarkers of treatment response. Treatment decisions are based on high-risk features such as advanced TNM stage and histology. The role of the tumour microenvironment, which can influence tumour progression and treatment response, has generated considerable interest. Patient-derived explant cultures allow preservation of native tissue architecture and tumour microenvironment. The aim of the scoping review is to evaluate the utility of patient-derived explant cultures as a preclinical model in colorectal cancer. METHODS A search was conducted using Ovid MEDLINE, EMBASE, Web of Science, and Cochrane databases from start of database records to September 1, 2022. We included all peer-reviewed human studies in English language which used patient-derived explants as a preclinical model in primary colorectal cancer. Eligible studies were grouped into the following categories: assessing model feasibility; exploring tumour microenvironment; assessing ex vivo drug responses; discovering and validating biomarkers. RESULTS A total of 60 studies were eligible. Fourteen studies demonstrated feasibility of using patient-derived explants as a preclinical model. Ten studies explored the tumour microenvironment. Thirty-eight studies assessed ex vivo drug responses of chemotherapy agents and targeted therapies. Twenty-four studies identified potential biomarkers of treatment response. CONCLUSIONS Given the preservation of tumour microenvironment and tumour heterogeneity, patient-derived explants has the potential to identify reliable biomarkers, treatment resistance mechanisms, and novel therapeutic agents. Further validation studies are required to characterise, refine and standardise this preclinical model before it can become a part of precision medicine in colorectal cancer.
Collapse
Affiliation(s)
- Milton Mui
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Molly Clark
- Department of Colorectal Surgery, Alfred Hospital, Melbourne, Victoria, Australia
| | - Tamara M S H Vu
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas Clemons
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
- Victorian Comprehensive Cancer Centre, The University of Melbourne Centre for Cancer Research, Melbourne, Victoria, Australia
| | - Sara Roth
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert Ramsay
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Michael
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alexander G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph C H Kong
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Colorectal Surgery, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Kayser C, Brauer A, Susanne S, Wandmacher AM. The challenge of making the right choice: patient avatars in the era of cancer immunotherapies. Front Immunol 2023; 14:1237565. [PMID: 37638045 PMCID: PMC10449253 DOI: 10.3389/fimmu.2023.1237565] [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: 06/09/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Immunotherapies are a key therapeutic strategy to fight cancer. Diverse approaches are used to activate tumor-directed immunity and to overcome tumor immune escape. The dynamic interplay between tumor cells and their tumor(immune)microenvironment (T(I)ME) poses a major challenge to create appropriate model systems. However, those model systems are needed to gain novel insights into tumor (immune) biology and a prerequisite to accurately develop and test immunotherapeutic approaches which can be successfully translated into clinical application. Several model systems have been established and advanced into so-called patient avatars to mimic the patient´s tumor biology. All models have their advantages but also disadvantages underscoring the necessity to pay attention in defining the rationale and requirements for which the patient avatar will be used. Here, we briefly outline the current state of tumor model systems used for tumor (immune)biological analysis as well as evaluation of immunotherapeutic agents. Finally, we provide a recommendation for further development to make patient avatars a complementary tool for testing and predicting immunotherapeutic strategies for personalization of tumor therapies.
Collapse
Affiliation(s)
- Charlotte Kayser
- Group of Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Annika Brauer
- Group of Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Sebens Susanne
- Group of Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Anna Maxi Wandmacher
- Group of Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
- Department of Internal Medicine II, University Hospital Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
5
|
Weitz J, Garg B, Martsinkovskiy A, Patel S, Tiriac H, Lowy AM. Pancreatic ductal adenocarcinoma induces neural injury that promotes a transcriptomic and functional repair signature by peripheral neuroglia. Oncogene 2023; 42:2536-2546. [PMID: 37433986 PMCID: PMC10880465 DOI: 10.1038/s41388-023-02775-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023]
Abstract
Perineural invasion (PNI) is the phenomenon whereby cancer cells invade the space surrounding nerves. PNI occurs frequently in epithelial malignancies, but is especially characteristic of pancreatic ductal adenocarcinoma (PDAC). The presence of PNI portends an increased incidence of local recurrence, metastasis and poorer overall survival. While interactions between tumor cells and nerves have been investigated, the etiology and initiating cues for PNI development is not well understood. Here, we used digital spatial profiling to reveal changes in the transcriptome and to allow for a functional analysis of neural-supportive cell types present within the tumor-nerve microenvironment of PDAC during PNI. We found that hypertrophic tumor-associated nerves within PDAC express transcriptomic signals of nerve damage including programmed cell death, Schwann cell proliferation signaling pathways, as well as macrophage clearance of apoptotic cell debris by phagocytosis. Moreover, we identified that neural hypertrophic regions have increased local neuroglial cell proliferation which was tracked using EdU tumor labeling in KPC mice, as well as frequent TUNEL positivity, suggestive of a high turnover rate. Functional calcium imaging studies using human PDAC organotypic slices confirmed nerve bundles had neuronal activity, as well as contained NGFR+ cells with high sustained calcium levels, which are indicative of apoptosis. This study reveals a common gene expression pattern that characterizes solid tumor-induced damage to local nerves. These data provide new insights into the pathobiology of the tumor-nerve microenvironment during PDAC as well as other gastrointestinal cancers.
Collapse
Affiliation(s)
- Jonathan Weitz
- Department of Surgery, University of California, San Diego, La Jolla, CA, CA 92093, USA.
| | - Bharti Garg
- Department of Surgery, University of California, San Diego, La Jolla, CA, CA 92093, USA
| | - Alexei Martsinkovskiy
- Department of Surgery, University of California, San Diego, La Jolla, CA, CA 92093, USA
| | - Sandip Patel
- Division of Hematology-Oncology in the Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Herve Tiriac
- Department of Surgery, University of California, San Diego, La Jolla, CA, CA 92093, USA
| | - Andrew M Lowy
- Department of Surgery, University of California, San Diego, La Jolla, CA, CA 92093, USA.
| |
Collapse
|
6
|
Siwczak F, Hiller C, Pfannkuche H, Schneider MR. Culture of vibrating microtome tissue slices as a 3D model in biomedical research. J Biol Eng 2023; 17:36. [PMID: 37264444 DOI: 10.1186/s13036-023-00357-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/21/2023] [Indexed: 06/03/2023] Open
Abstract
The basic idea behind the use of 3-dimensional (3D) tools in biomedical research is the assumption that the structures under study will perform at the best in vitro if cultivated in an environment that is as similar as possible to their natural in vivo embedding. Tissue slicing fulfills this premise optimally: it is an accessible, unexpensive, imaging-friendly, and technically rather simple procedure which largely preserves the extracellular matrix and includes all or at least most supportive cell types in the correct tissue architecture with little cellular damage. Vibrating microtomes (vibratomes) can further improve the quality of the generated slices because of the lateral, saw-like movement of the blade, which significantly reduces tissue pulling or tearing compared to a straight cut. In spite of its obvious advantages, vibrating microtome slices are rather underrepresented in the current discussion on 3D tools, which is dominated by methods as organoids, organ-on-chip and bioprinting. Here, we review the development of vibrating microtome tissue slices, the major technical features underlying its application, as well as its current use and potential advances, such as a combination with novel microfluidic culture chambers. Once fully integrated into the 3D toolbox, tissue slices may significantly contribute to decrease the use of laboratory animals and is likely to have a strong impact on basic and translational research as well as drug screening.
Collapse
Affiliation(s)
- Fatina Siwczak
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Charlotte Hiller
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Helga Pfannkuche
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Marlon R Schneider
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany.
| |
Collapse
|
7
|
Wu KZ, Adine C, Mitriashkin A, Aw BJJ, Iyer NG, Fong ELS. Making In Vitro Tumor Models Whole Again. Adv Healthc Mater 2023; 12:e2202279. [PMID: 36718949 PMCID: PMC11469124 DOI: 10.1002/adhm.202202279] [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: 09/06/2022] [Revised: 01/04/2023] [Indexed: 02/01/2023]
Abstract
As a reductionist approach, patient-derived in vitro tumor models are inherently still too simplistic for personalized drug testing as they do not capture many characteristics of the tumor microenvironment (TME), such as tumor architecture and stromal heterogeneity. This is especially problematic for assessing stromal-targeting drugs such as immunotherapies in which the density and distribution of immune and other stromal cells determine drug efficacy. On the other end, in vivo models are typically costly, low-throughput, and time-consuming to establish. Ex vivo patient-derived tumor explant (PDE) cultures involve the culture of resected tumor fragments that potentially retain the intact TME of the original tumor. Although developed decades ago, PDE cultures have not been widely adopted likely because of their low-throughput and poor long-term viability. However, with growing recognition of the importance of patient-specific TME in mediating drug response, especially in the field of immune-oncology, there is an urgent need to resurrect these holistic cultures. In this Review, the key limitations of patient-derived tumor explant cultures are outlined and technologies that have been developed or could be employed to address these limitations are discussed. Engineered holistic tumor explant cultures may truly realize the concept of personalized medicine for cancer patients.
Collapse
Affiliation(s)
- Kenny Zhuoran Wu
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Christabella Adine
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Aleksandr Mitriashkin
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Benjamin Jun Jie Aw
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - N. Gopalakrishna Iyer
- Department of Head and Neck Surgery, Division of Surgery and Surgical OncologyDuke‐NUS Medical SchoolSingapore169857Singapore
- Department of Head and Neck SurgeryNational Cancer Centre SingaporeSingapore169610Singapore
| | - Eliza Li Shan Fong
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
- The N.1 Institute for HealthNational University of SingaporeSingapore117456Singapore
- Cancer Science Institute (CSI)National University of SingaporeSingapore117599Singapore
| |
Collapse
|
8
|
Salas A, García-García P, Díaz-Rodríguez P, Évora C, Almeida TA, Delgado A. New local ganirelix sustained release therapy for uterine leiomyoma. Evaluation in a preclinical organ model. Biomed Pharmacother 2022; 156:113909. [DOI: 10.1016/j.biopha.2022.113909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/02/2022] Open
|
9
|
Genta S, Coburn B, Cescon DW, Spreafico A. Patient-derived cancer models: Valuable platforms for anticancer drug testing. Front Oncol 2022; 12:976065. [PMID: 36033445 PMCID: PMC9413077 DOI: 10.3389/fonc.2022.976065] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Molecularly targeted treatments and immunotherapy are cornerstones in oncology, with demonstrated efficacy across different tumor types. Nevertheless, the overwhelming majority metastatic disease is incurable due to the onset of drug resistance. Preclinical models including genetically engineered mouse models, patient-derived xenografts and two- and three-dimensional cell cultures have emerged as a useful resource to study mechanisms of cancer progression and predict efficacy of anticancer drugs. However, variables including tumor heterogeneity and the complexities of the microenvironment can impair the faithfulness of these platforms. Here, we will discuss advantages and limitations of these preclinical models, their applicability for drug testing and in co-clinical trials and potential strategies to increase their reliability in predicting responsiveness to anticancer medications.
Collapse
Affiliation(s)
- Sofia Genta
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Bryan Coburn
- Division of Infectious Diseases, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - David W. Cescon
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Lim CY, Chang JH, Lee WS, Kim J, Park IY. CD40 Agonists Alter the Pancreatic Cancer Microenvironment by Shifting the Macrophage Phenotype toward M1 and Suppress Human Pancreatic Cancer in Organotypic Slice Cultures. Gut Liver 2022; 16:645-659. [PMID: 34933280 PMCID: PMC9289829 DOI: 10.5009/gnl210311] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/24/2021] [Accepted: 09/03/2021] [Indexed: 11/04/2022] Open
Abstract
Background/Aims CD40 agonists are thought to generate antitumor effects on pancreatic cancer via macrophages and T cells. We aimed to investigate the role of CD40 agonists in the differentiation of macrophages and treatment of human pancreatic adenocarcinoma. Methods Immunohistochemistry was performed on paraffin-embedded surgical blocks from patients with pancreatic cancers to evaluate macrophage phenotypes and their relationship with survival. The effects of CD40 agonists on macrophage phenotypes and human pancreatic cancer were evaluated utilizing cell cocultures and organotypic slice cultures. Results CD163+ (predominant in M2 macrophages) and FOXP3+ (predominant in regulatory T cells) expression levels in the tumors were significantly lower in patients with stage IB pancreatic cancer than in those with stage II or III disease (p=0.002 and p=0.003, respectively). Patients with high CD163+ expression had shorter overall survival than those with low CD163+ expression (p=0.002). In vitro treatment of THP-1 macrophages with a CD40 agonist led to an increase in HLA-DR+ (predominant in M1 macrophages) and a decrease in CD163+ expression in THP-1 cells. Cell cocultures showed that CD40 agonists facilitate the suppression of PANC-1 human pancreatic cancer cells by THP-1 macrophages. Organotypic slice cultures showed that CD40 agonists alter the pancreatic cancer microenvironment by shifting the macrophage phenotype toward M1 (increase HLA-DR+ and decrease CD163+ expression), decreasing the abundance of regulatory T cells, and increasing tumor cell apoptosis. Conclusions CD163 is related to advanced human pancreatic cancer stages and shorter overall survival. CD40 agonists alter macrophage phenotype polarization to favor the M1 phenotype and suppress human pancreatic cancer.
Collapse
Affiliation(s)
- Chae Yoon Lim
- Institute of Clinical Medicine Research, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Hyuck Chang
- Departments of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Won Sun Lee
- Institute of Clinical Medicine Research, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeana Kim
- Departments of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Il Young Park
- Departments of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
11
|
Principe DR, Aissa AF, Kumar S, Pham TND, Underwood PW, Nair R, Ke R, Rana B, Trevino JG, Munshi HG, Benevolenskaya EV, Rana A. Calcium channel blockers potentiate gemcitabine chemotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:e2200143119. [PMID: 35476525 PMCID: PMC9170157 DOI: 10.1073/pnas.2200143119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/19/2022] [Indexed: 12/15/2022] Open
Abstract
There is currently no effective treatment for pancreatic ductal adenocarcinoma (PDAC). While palliative chemotherapy offers a survival benefit to most patients, nearly all will eventually progress on treatment and long-term survivability remains poor. Given the lack of subsequent line treatment options, in this study, we sought to identify novel strategies to prevent, delay, or overcome resistance to gemcitabine, one of the most widely used medications in PDAC. Using a combination of single-cell RNA sequencing and high-throughput proteomic analysis, we identified a subset of gemcitabine-resistant tumor cells enriched for calcium/calmodulin signaling. Pharmacologic inhibition of calcium-dependent calmodulin activation led to the rapid loss of drug-resistant phenotypes in vitro, which additional single-cell RNA sequencing identified was due to impaired activation of the RAS/ERK signaling pathway. Consistent with these observations, calcium chelation or depletion of calcium in the culture media also impaired ERK activation in gemcitabine-resistant cells, and restored therapeutic responses to gemcitabine in vitro. We observed similar results using calcium channel blockers (CCBs) such as amlodipine, which inhibited prosurvival ERK signaling in vitro and markedly enhanced therapeutic responses to gemcitabine in both orthotopic xenografts and transgenic models of PDAC. Combined, these results offer insight into a potential means of gemcitabine resistance and suggest that select CCBs may provide a clinical benefit to PDAC patients receiving gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Alexandre F. Aissa
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Sandeep Kumar
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
| | - Patrick W. Underwood
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32611
| | - Rakesh Nair
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Rong Ke
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Basabi Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Jose G. Trevino
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University, Richmond, VA 23284
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
- Jesse Brown VA Medical Center, Chicago, IL 60612
| | | | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Jesse Brown VA Medical Center, Chicago, IL 60612
| |
Collapse
|
12
|
XP-524 is a dual-BET/EP300 inhibitor that represses oncogenic KRAS and potentiates immune checkpoint inhibition in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:2116764119. [PMID: 35064087 PMCID: PMC8795568 DOI: 10.1073/pnas.2116764119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
There are currently no effective treatments for pancreatic ductal adenocarcinoma (PDAC), which displays widespread resistance to chemotherapy, radiation therapy, and immunotherapy. Here, we demonstrate that the multispecificity BET/EP300 inhibitor XP-524 has pronounced single-agent efficacy in vitro, in vivo, and in ex vivo human PDAC slice cultures, functioning in part by attenuating oncogenic KRAS signaling. In vivo XP-524 led to extensive reprogramming of the pancreatic tumor microenvironment, sensitizing murine carcinoma to immune checkpoint inhibition and further extending survival. Given the urgent need for therapeutic approaches in PDAC, the combination of XP-524 and immune checkpoint inhibition warrants additional exploration. Pancreatic ductal adenocarcinoma (PDAC) is associated with extensive dysregulation of the epigenome and epigenetic regulators, such as bromodomain and extraterminal motif (BET) proteins, have been suggested as potential targets for therapy. However, single-agent BET inhibition has shown poor efficacy in clinical trials, and no epigenetic approaches are currently used in PDAC. To circumvent the limitations of the current generation of BET inhibitors, we developed the compound XP-524 as an inhibitor of the BET protein BRD4 and the histone acetyltransferase EP300/CBP, both of which are ubiquitously expressed in PDAC tissues and cooperate to enhance tumorigenesis. XP-524 showed increased potency and superior tumoricidal activity than the benchmark BET inhibitor JQ-1 in vitro, with comparable efficacy to higher-dose JQ-1 combined with the EP300/CBP inhibitor SGC-CBP30. We determined that this is in part due to the epigenetic silencing of KRAS in vitro, with similar results observed using ex vivo slice cultures of human PDAC tumors. Accordingly, XP-524 prevented KRAS-induced, neoplastic transformation in vivo and extended survival in two transgenic mouse models of aggressive PDAC. In addition to the inhibition of KRAS/MAPK signaling, XP-524 also enhanced the presentation of self-peptide and tumor recruitment of cytotoxic T lymphocytes, though these lymphocytes remained refractory from full activation. We, therefore, combined XP-524 with an anti–PD-1 antibody in vivo, which reactivated the cytotoxic immune program and extended survival well beyond XP-524 in monotherapy. Pending a comprehensive safety evaluation, these results suggest that XP-524 may benefit PDAC patients and warrant further exploration, particularly in combination with immune checkpoint inhibition.
Collapse
|
13
|
Templeton AR, Jeffery PL, Thomas PB, Perera MPJ, Ng G, Calabrese AR, Nicholls C, Mackenzie NJ, Wood J, Bray LJ, Vela I, Thompson EW, Williams ED. Patient-Derived Explants as a Precision Medicine Patient-Proximal Testing Platform Informing Cancer Management. Front Oncol 2022; 11:767697. [PMID: 34988013 PMCID: PMC8721047 DOI: 10.3389/fonc.2021.767697] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Precision medicine approaches that inform clinical management of individuals with cancer are progressively advancing. Patient-derived explants (PDEs) provide a patient-proximal ex vivo platform that can be used to assess sensitivity to standard of care (SOC) therapies and novel agents. PDEs have several advantages as a patient-proximal model compared to current preclinical models, as they maintain the phenotype and microenvironment of the individual tumor. However, the longevity of PDEs is not compatible with the timeframe required to incorporate candidate therapeutic options identified by whole exome sequencing (WES) of the patient’s tumor. This review investigates how PDE longevity varies across tumor streams and how this is influenced by tissue preparation. Improving longevity of PDEs will enable individualized therapeutics testing, and thus contribute to improving outcomes for people with cancer.
Collapse
Affiliation(s)
- Abby R Templeton
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Penny L Jeffery
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Patrick B Thomas
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Mahasha P J Perera
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia.,Department of Urology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Gary Ng
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Department of Medical Oncology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Alivia R Calabrese
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Clarissa Nicholls
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Nathan J Mackenzie
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Jack Wood
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Laura J Bray
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Ian Vela
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia.,Department of Urology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Erik W Thompson
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
14
|
Weitz JR, Tiriac H, Hurtado de Mendoza T, Wascher A, Lowy AM. Using Organotypic Tissue Slices to Investigate the Microenvironment of Pancreatic Cancer: Pharmacotyping and Beyond. Cancers (Basel) 2021; 13:cancers13194991. [PMID: 34638476 PMCID: PMC8507648 DOI: 10.3390/cancers13194991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) has the highest mortality rate of all major cancers and, disappointingly, neither immune- nor stroma-directed therapies are found to improve upon the current standard of care. Among the most challenging aspects of PDAC biology which impede clinical success are the physiological features of the pancreatic cancer microenvironment (TME), including the presence of a highly fibrotic extracellular matrix marked by perineural invasion and an immunosuppressive milieu. Many current strategies for PDAC therapy are focused on altering these features to improve therapeutic efficacy. This review discusses the recent investigations using organotypic tumor slices as a model system to study cellular and extracellular interactions of the pancreatic TME. Future studies utilizing such models may provide new insights into the TME by identifying mechanisms of communication between multiple cell types and investigating novel therapeutic approaches for personalized cancer therapy. Abstract Organotypic tissue slices prepared from patient tumors are a semi-intact ex vivo preparation that recapitulates many aspects of the tumor microenvironment (TME). While connections to the vasculature and nervous system are severed, the integral functional elements of the tumor remain intact for many days during the slice culture. During this window of time, the slice platforms offer a suite of molecular, biomechanical and functional tools to investigate PDAC biology. In this review, we first briefly discuss the development of pancreatic tissue slices as a model system. Next, we touch upon using slices as an orthogonal approach to study the TME as compared to other established 3D models, such as organoids. Distinct from most other models, the pancreatic slices contain autologous immune and other stromal cells. Taking advantage of the existing immune cells within the slices, we will discuss the breakthrough studies which investigate the immune compartment in the pancreas slices. These studies will provide an important framework for future investigations seeking to exploit or reprogram the TME for cancer therapy.
Collapse
Affiliation(s)
- Jonathan Robert Weitz
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; (J.R.W.); (H.T.); (T.H.d.M.); (A.W.)
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Herve Tiriac
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; (J.R.W.); (H.T.); (T.H.d.M.); (A.W.)
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Tatiana Hurtado de Mendoza
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; (J.R.W.); (H.T.); (T.H.d.M.); (A.W.)
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alexis Wascher
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; (J.R.W.); (H.T.); (T.H.d.M.); (A.W.)
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Andrew M. Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; (J.R.W.); (H.T.); (T.H.d.M.); (A.W.)
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
- Correspondence: ; Tel.: +1-858-822-2124
| |
Collapse
|
15
|
Evaluation of the Chemotherapy Drug Response Using Organotypic Cultures of Osteosarcoma Tumours from Mice Models and Canine Patients. Cancers (Basel) 2021; 13:cancers13194890. [PMID: 34638373 PMCID: PMC8507898 DOI: 10.3390/cancers13194890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Osteosarcoma is a bone cancer with 75% of cases occurring in people younger than 25 years old. 35–45% of patients demonstrate resistance to chemotherapeutics and critically, survival rates for osteosarcoma is only 10–30% for patients with metastases. Therefore, reliable and patient-specific drug testing modalities are needed. Organotypic slice culture consists of sections of tumours, which survive and preserve the tumours mechanical and cellular properties, thereby enabling personalised testing of drugs. This study aimed to characterise organotypic slice cultures of osteosarcoma bone tumours derived from mice and dogs and to use these models for testing of anti-tumoural drugs. This study reports the various cell constituents of the model and the maintenance of osteosarcoma organotypic cultures over several weeks. A significantly decreased sensitivity to chemotherapy in 3D organotypic culture relative to 2D monolayer was found, highlighting the need to test anti-cancer drugs in a more personalized and biomimetic manner. Abstract Improvements in the clinical outcome of osteosarcoma have plateaued in recent decades with poor translation between preclinical testing and clinical efficacy. Organotypic cultures retain key features of patient tumours, such as a myriad of cell types organized within an extracellular matrix, thereby presenting a more realistic and personalised screening of chemotherapeutic agents ex vivo. To test this concept for the first time in osteosarcoma, murine and canine osteosarcoma organotypic models were maintained for up to 21 days and in-depth analysis identified proportions of immune and stromal cells present at levels comparable to that reported in vivo in the literature. Cytotoxicity testing of a range of chemotherapeutic drugs (mafosfamide, cisplatin, methotrexate, etoposide, and doxorubicin) on murine organotypic culture ex vivo found limited response to treatment, with immune and stromal cells demonstrating enhanced survival over the global tumour cell population. Furthermore, significantly decreased sensitivity to a range of chemotherapeutics in 3D organotypic culture relative to 2D monolayer was observed, with subsequent investigation confirming reduced sensitivity in 3D than in 2D, even at equivalent levels of drug uptake. Finally, as proof of concept for the application of this model to personalised drug screening, chemotherapy testing with doxorubicin was performed on biopsies obtained from canine osteosarcoma patients. Together, this study highlights the importance of recapitulating the 3D tumour multicellular microenvironment to better predict drug response and provides evidence for the utility and possibilities of organotypic culture for enhanced preclinical selection and evaluation of chemotherapeutics targeting osteosarcoma.
Collapse
|
16
|
Hußtegge M, Hoang NA, Rebstock J, Monecke A, Gockel I, Weimann A, Schumacher G, Bechmann I, Lordick F, Kallendrusch S, Körfer J. PD-1 inhibition in patient derived tissue cultures of human gastric and gastroesophageal adenocarcinoma. Oncoimmunology 2021; 10:1960729. [PMID: 34434611 PMCID: PMC8381835 DOI: 10.1080/2162402x.2021.1960729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Emerging immunotherapies quest for better patient stratification in cancer treatment decisions. Moderate response rates of PD-1 inhibition in gastric and esophagogastric junction cancers urge for meaningful human model systems that allow for investigating immune responses ex vivo. Here, the standardized patient-derived tissue culture (PDTC) model was applied to investigate tumor response to the PD-1 inhibitor Nivolumab and the CD3/CD28 t-lymphocyte activator ImmunoCultTM. Resident t-lymphocytes, tumor proliferation and apoptosis, as well as bulk gene expression data were analyzed after 72 h of PD-1 inhibition either as monotherapy or combined with Oxaliplatin or ImmunoCultTM. Individual responses to PD-1 inhibition were found ex vivo and combination with chemotherapy or t-lymphocyte activation led to enhanced antitumoral effects in PDTCs. T-lymphocyte activation as well as the addition of pre-cultured peripheral blood mononuclear cells improved PDTC for studying t-lymphocyte and tumor cell communication. These data support the potential of PDTC to investigate immunotherapy ex vivo in gastric and esophagogastric junction cancer.
Collapse
Affiliation(s)
- Marlon Hußtegge
- Institute of Anatomy, University of Leipzig, Leipzig.,Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Ngoc Anh Hoang
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Jakob Rebstock
- Institute of Anatomy, University of Leipzig, Leipzig.,Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Astrid Monecke
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Ines Gockel
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Arved Weimann
- Department for General and Visceral Surgery, Hospital St. Georg Leipzig, Leipzig, Germany
| | - Guido Schumacher
- Department for General and Visceral Surgery, Hospital Braunschweig, Braunschweig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig
| | - Florian Lordick
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | | | - Justus Körfer
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Supadmanaba IGP, Comandatore A, Morelli L, Giovannetti E, Lagerweij T. Organotypic-liver slide culture systems to explore the role of extracellular vesicles in pancreatic cancer metastatic behavior and guide new therapeutic approaches. Expert Opin Drug Metab Toxicol 2021; 17:937-946. [PMID: 33945374 DOI: 10.1080/17425255.2021.1925646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/30/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Recent studies suggested that extracellular vesicles (EVs) play a role both in the metastatic niche formation and in the progression of several tumors, including pancreatic cancer. In particular, the effects of EVs on metastasis should be studied in model systems that take into account both the tumor cells and the metastatic site/tumor microenvironment. Studies with labeled EVs or EV-secreting cells in ex vivo models will reflect the physiological and pathological functions of EVs. The organotypic-tissue slide culture systems can fulfill such a role.Areas covered: This review provides an overview of available organotypic-culture slide systems. We specifically focus on the assay system of liver culture-slides in combination with pancreatic tumors, which can be modulated to test the efficacy of new therapeutic approaches.Expert opinion: The intercellular exchange of EVs has emerged as a biologically relevant phenomenon to drive cancer metastasis. However, further models need to be developed to better elucidate the functional roles of EVs. The use of novel organotypic slide culture systems provides the opportunity to explore the role of EVs in the metastatic behavior of pancreatic cancer, decreasing the use of costly and cumbersome organoid or animal models.
Collapse
Affiliation(s)
- I Gede Putu Supadmanaba
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Biochemistry Department, Faculty of Medicine, Universitas Udayana, Denpasar, Bali, Indonesia
| | - Annalisa Comandatore
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Tonny Lagerweij
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Domínguez-Bendala J, Qadir MMF, Pastori RL. Temporal single-cell regeneration studies: the greatest thing since sliced pancreas? Trends Endocrinol Metab 2021; 32:433-443. [PMID: 34006411 PMCID: PMC8239162 DOI: 10.1016/j.tem.2021.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023]
Abstract
The application of single-cell analytic techniques to the study of stem/progenitor cell niches supports the emerging view that pancreatic cell lineages are in a state of flux between differentiation stages. For all their value, however, such analyses merely offer a snapshot of the cellular palette of the tissue at any given time point. Conclusions about potential developmental/regeneration paths are solely based on bioinformatics inferences. In this context, the advent of new techniques for the long-term culture and lineage tracing of human pancreatic slices offers a virtual window into the native organ and presents the field with a unique opportunity to serially resolve pancreatic regeneration dynamics at the single-cell level.
Collapse
Affiliation(s)
- Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mirza Muhammad Fahd Qadir
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
19
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
20
|
FOXM1 Inhibition in Ovarian Cancer Tissue Cultures Affects Individual Treatment Susceptibility Ex Vivo. Cancers (Basel) 2021; 13:cancers13050956. [PMID: 33668819 PMCID: PMC7956612 DOI: 10.3390/cancers13050956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Late diagnosis of ovarian cancer is a major reason for the high mortality rate of this tumor entity. The time to determine tumor susceptibility to treatment is scarce and resistance to therapy occurs very frequently. Here, we aim for a model system that can determine tumor response to (I) study novel drugs and (II) enhance patient stratification. Tissue specimens (n = 10) were acquired from fresh surgical samples. Tissue cultures were cultivated and treated with clinically relevant therapeutics and an FOXM1 inhibitor for 3–6 days. The transcription factor FOXM1 is a key regulator of tumor survival affecting multiple cancerogenic target genes. Gene expression of FOXM1 and its targets BRCA1/2 and RAD51 were investigated together with tumor susceptibility. Tissue cultures successfully demonstrated the individual benefit of FOXM1 inhibition and revealed the potency of the complex model system for oncological research. Abstract Diagnosis in an advanced state is a major hallmark of ovarian cancer and recurrence after first line treatment is common. With upcoming novel therapies, tumor markers that support patient stratification are urgently needed to prevent ineffective therapy. Therefore, the transcription factor FOXM1 is a promising target in ovarian cancer as it is frequently overexpressed and associated with poor prognosis. In this study, fresh tissue specimens of 10 ovarian cancers were collected to investigate tissue cultures in their ability to predict individual treatment susceptibility and to identify the benefit of FOXM1 inhibition. FOXM1 inhibition was induced by thiostrepton (3 µM). Carboplatin (0.2, 2 and 20 µM) and olaparib (10 µM) were applied and tumor susceptibility was analyzed by tumor cell proliferation and apoptosis in immunofluorescence microscopy. Resistance mechanisms were investigated by determining the gene expression of FOXM1 and its targets BRCA1/2 and RAD51. Ovarian cancer tissue was successfully maintained for up to 14 days ex vivo, preserving morphological characteristics of the native specimen. Thiostrepton downregulated FOXM1 expression in tissue culture. Individual responses were observed after combined treatment with carboplatin or olaparib. Thus, we successfully implemented a complex tissue culture model to ovarian cancer and showed potential benefit of combined FOXM1 inhibition.
Collapse
|
21
|
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: 1.5] [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
|
22
|
Gu ZT, Li ZZ, Wang CF. Research advances of intracellular mechanisms underlying gemcitabine resistance in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2020; 28:1150-1161. [DOI: 10.11569/wcjd.v28.i22.1150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most deadly malignant tumors that endanger human health, and pancreatic ductal adenocarcinoma (PDAC) is the most common histological type. Due to the lack of specific clinical symptoms, physical signs, and effective screening biomarkers for early stage PDAC, only 15%-20% of patients are qualified for surgical resection. Consequently, gemcitabine (GEM)-based monotherapy or combination therapy is still the most important or even the only treatment option. However, the overall response rate of PDAC to GEM is less than 20%, and GEM resistance is one of the most important factors affecting the efficacy of chemotherapy. At present, the mechanism of GEM resistance has not been clarified, which may involve congenital and acquired regulation. The heterogeneity of PDAC further increases its complexity. However, regulation of intracellular signaling pathways is the ultimate event to induce GEM resistance. This article will review the recent advances in research of GEM metabolism and regulation of signaling pathways in PDAC cells, and discuss potential GEM chemosensitization strategies, in order to improve the effective rate of chemotherapy and the outcome.
Collapse
Affiliation(s)
- Zong-Ting Gu
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zong-Ze Li
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | |
Collapse
|
23
|
Patient-derived tumour models for personalized therapeutics in urological cancers. Nat Rev Urol 2020; 18:33-45. [PMID: 33173206 DOI: 10.1038/s41585-020-00389-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2020] [Indexed: 12/24/2022]
Abstract
Preclinical knowledge of dysregulated pathways and potential biomarkers for urological cancers has undergone limited translation into the clinic. Moreover, the low approval rate of new anticancer drugs and the heterogeneous drug responses in patients indicate that current preclinical models do not always reflect the complexity of malignant disease. Patient-derived tumour models used in preclinical uro-oncology research include 3D culture systems, organotypic tissue slices and patient-derived xenograft models. Technological innovations have enabled major improvements in the capacity of these tumour models to reproduce the clinical complexity of urological cancers. Each type of patient-derived model has inherent advantages and limitations that can be exploited, either alone or in combination, to gather specific knowledge on clinical challenges and address unmet clinical needs. Nevertheless, few opportunities exist for patients with urological cancers to benefit from personalized therapeutic approaches. Clinical validation of experimental data is needed to facilitate the translation and implementation of preclinical knowledge into treatment decision making.
Collapse
|
24
|
Solanki A, King D, Thibault G, Wang L, Gibbs SL. Quantification of fluorophore distribution and therapeutic response in matched in vivo and ex vivo pancreatic cancer model systems. PLoS One 2020; 15:e0229407. [PMID: 32097436 PMCID: PMC7041865 DOI: 10.1371/journal.pone.0229407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/05/2020] [Indexed: 12/18/2022] Open
Abstract
Therapeutic resistance plagues cancer outcomes, challenging treatment particularly in aggressive disease. A unique method to decipher drug interactions with their targets and inform therapy is to employ fluorescence-based screening tools; however, to implement productive screening assays, adequate model systems must be developed. Patient-derived pancreatic cancer models (e.g., cell culture, patient-derived xenograft mouse models, and organoids) have been traditionally utilized to predict personalized therapeutic response. However, cost, long read out times and the inability to fully recapitulate the tumor microenvironment have rendered most models incompatible with clinical decision making for pancreatic ductal adenocarcinoma (PDAC) patients. Tumor explant cultures, where patient tissue can be kept viable for up to weeks, have garnered interest as a platform for delivering personalized therapeutic prediction on a clinically relevant timeline. To fully explore this ex vivo platform, a series of studies were completed to quantitatively compare in vivo models with tumor explants, examining gemcitabine therapeutic efficacy, small molecule uptake and drug-target engagement using a novel fluorescently-labeled gemcitabine conjugate. This initial work shows promise for patient-specific therapeutic selection, where tumor explant drug distribution and response recapitulated the in vivo behavior and could provide a valuable platform for understanding mechanisms of therapeutic response and resistance.
Collapse
Affiliation(s)
- Allison Solanki
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Diana King
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Guillaume Thibault
- Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lei Wang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Summer L. Gibbs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
25
|
Bray LJ, Hutmacher DW, Bock N. Addressing Patient Specificity in the Engineering of Tumor Models. Front Bioeng Biotechnol 2019; 7:217. [PMID: 31572718 PMCID: PMC6751285 DOI: 10.3389/fbioe.2019.00217] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is challenged by the heterogeneous nature of cancer, where prognosis depends on tumor type and disease stage, as well as previous treatments. Optimal patient stratification is critical for the development and validation of effective treatments, yet pre-clinical model systems are lacking in the delivery of effective individualized platforms that reflect distinct patient-specific clinical situations. Advances in cancer cell biology, biofabrication, and microengineering technologies have led to the development of more complex in vitro three-dimensional (3D) models to act as drug testing platforms and to elucidate novel cancer mechanisms. Mostly, these strategies have enabled researchers to account for the tumor microenvironment context including tumor-stroma interactions, a key factor of heterogeneity that affects both progression and therapeutic resistance. This is aided by state-of-the-art biomaterials and tissue engineering technologies, coupled with reproducible and high-throughput platforms that enable modeling of relevant physical and chemical factors. Yet, the translation of these models and technologies has been impaired by neglecting to incorporate patient-derived cells or tissues, and largely focusing on immortalized cell lines instead, contributing to drug failure rates. While this is a necessary step to establish and validate new models, a paradigm shift is needed to enable the systematic inclusion of patient-derived materials in the design and use of such models. In this review, we first present an overview of the components responsible for heterogeneity in different tumor microenvironments. Next, we introduce the state-of-the-art of current in vitro 3D cancer models employing patient-derived materials in traditional scaffold-free approaches, followed by novel bioengineered scaffold-based approaches, and further supported by dynamic systems such as bioreactors, microfluidics, and tumor-on-a-chip devices. We critically discuss the challenges and clinical prospects of models that have succeeded in providing clinical relevance and impact, and present emerging concepts of novel cancer model systems that are addressing patient specificity, the next frontier to be tackled by the field.
Collapse
Affiliation(s)
- Laura J. Bray
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Dietmar W. Hutmacher
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
- Australian Research Council (ARC) Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
26
|
Antitumoral Effect of Laurinterol on 3D Culture of Breast Cancer Explants. Mar Drugs 2019; 17:md17040201. [PMID: 30934912 PMCID: PMC6520734 DOI: 10.3390/md17040201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/18/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
Macroalgae represent an important source of bioactive compounds with a wide range of biotechnological applications. Overall, the discovery of effective cytotoxic compounds with pharmaceutical potential is a significant challenge, mostly because they are scarce in nature or their total synthesis is not efficient, while the bioprospecting models currently used do not predict clinical responses. Given this context, we used three-dimensional (3D) cultures of human breast cancer explants to evaluate the antitumoral effect of laurinterol, the major compound of an ethanolic extract of Laurencia johnstonii. To this end, we evaluated the metabolic and histopathological effects of the crude extract of L. johnstonii and laurinterol on Vero and MCF-7 cells, in addition to breast cancer explants. We observed a dose-dependent inhibition of the metabolic activity, as well as morphologic and nuclear changes characteristic of apoptosis. On the other hand, a reduced metabolic viability and marked necrosis areas were observed in breast cancer explants incubated with the crude extract, while explants treated with laurinterol exhibited a heterogeneous response which was associated with the individual response of each human tumor sample. This study supports the cytotoxic and antitumoral effects of laurinterol in in vitro cell cultures and in ex vivo organotypic cultures of human breast cancer explants.
Collapse
|