1
|
Pecci V, Borsa M, Aiello A, De Martino S, Cis L, Ripoli C, Rotili D, Pierconti F, Pinto F, Grassi C, Gaetano C, Farsetti A, Nanni S. Bromodomain and Extra-Terminal Family Proteins BRD2, BRD3, and BRD4 Contribute to H19-Dependent Transcriptional Regulation of Cell Adhesion Molecules, Modulating Metastatic Dissemination Program in Prostate Cancer. Noncoding RNA 2025; 11:33. [PMID: 40407591 PMCID: PMC12101203 DOI: 10.3390/ncrna11030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic prostate cancer (PCa) remains a major clinical challenge with limited therapeutic options. The long non-coding RNA H19 has been implicated in regulating cell adhesion molecules and collective migration, key features of metastatic dissemination. This study investigates the role of the Bromodomain and Extra-Terminal (BET) proteins BRD2, BRD3, and BRD4 in the H19-dependent transcriptional regulation of cell adhesion molecules. Currently, the major effects of BET inhibitors require androgen receptor (AR) expression. METHODS H19 was stably silenced in PC-3 (AR-null) and 22Rv1 (AR-positive) castration-resistant PCa cells. The cells were treated with the pan-BET inhibitors JQ1 and OTX015 or the BET degrader dBET6. In vivo, the effects of JQ1 were evaluated in xenograft mouse models. Chromatin immunoprecipitation (ChIP) and RNA-ChIP were used to assess BET protein recruitment and interaction with cell adhesion gene loci and H19. Organotypic slice cultures (OSCs) from fresh PCa surgical specimens were used as ex vivo models to validate transcriptional changes and BRD4 recruitment. RESULTS BET inhibition significantly reduced the expression of β4 integrin and E-cadherin and cell proliferation in both basal conditions, and following H19 knockdown in PC-3 and 22Rv1 cells. These effects were mirrored in JQ1-treated tumor xenografts, which showed marker downregulation and tumor regression. ChIP assays revealed that BRD4, more than BRD2/3, was enriched on β4 integrin and E-cadherin promoters, especially in regions marked by H3K27ac. H19 silencing markedly enhanced BRD4 promoter occupancy. RNA-ChIP confirmed a specific interaction between BRD4 and H19. These findings were validated in OSCs, reinforcing their clinical relevance. CONCLUSIONS Our study demonstrates that BRD4 epigenetically regulates the H19-mediated transcriptional control of adhesion molecules involved in collective migration and metastatic dissemination. Importantly, these effects are independent of AR status, suggesting that targeting the H19/BRD4 axis may represent a promising therapeutic avenue for advanced PCa.
Collapse
Affiliation(s)
- Valeria Pecci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (M.B.); (L.C.)
| | - Melissa Borsa
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (M.B.); (L.C.)
| | - Aurora Aiello
- National Research Council (CNR)-IASI, 00185 Rome, Italy; (A.A.); (S.D.M.)
| | - Sara De Martino
- National Research Council (CNR)-IASI, 00185 Rome, Italy; (A.A.); (S.D.M.)
| | - Luca Cis
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (M.B.); (L.C.)
- National Research Council (CNR)-IASI, 00185 Rome, Italy; (A.A.); (S.D.M.)
| | - Cristian Ripoli
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (C.R.); (F.P.); (F.P.); (C.G.)
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Dante Rotili
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Francesco Pierconti
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (C.R.); (F.P.); (F.P.); (C.G.)
- Department of Woman, Child and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Pinto
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (C.R.); (F.P.); (F.P.); (C.G.)
| | - Claudio Grassi
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (C.R.); (F.P.); (F.P.); (C.G.)
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Antonella Farsetti
- National Research Council (CNR)-IASI, 00185 Rome, Italy; (A.A.); (S.D.M.)
| | - Simona Nanni
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (M.B.); (L.C.)
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (C.R.); (F.P.); (F.P.); (C.G.)
| |
Collapse
|
2
|
Steindl A, Valiente M. Potential of ex vivo organotypic slice cultures in neuro-oncology. Neuro Oncol 2025; 27:338-351. [PMID: 39504579 PMCID: PMC11812025 DOI: 10.1093/neuonc/noae195] [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] [Indexed: 11/08/2024] Open
Abstract
Over recent decades, in vitro and in vivo models have significantly advanced brain cancer research; however, each presents distinct challenges for accurately mimicking in situ conditions. In response, organotypic slice cultures have emerged as a promising model recapitulating precisely specific in vivo phenotypes through an ex vivo approach. Ex vivo organotypic brain slice models can integrate biological relevance and patient-specific variability early in drug discovery, thereby aiming for more precise treatment stratification. However, the challenges of obtaining representative fresh brain tissue, ensuring reproducibility, and maintaining essential central nervous system (CNS)-specific conditions reflecting the in situ situation over time have limited the direct application of ex vivo organotypic slice cultures in robust clinical trials. In this review, we explore the benefits and possible limitations of ex vivo organotypic brain slice cultures in neuro-oncological research. Additionally, we share insights from clinical experts in neuro-oncology on how to overcome these current limitations and improve the practical application of organotypic brain slice cultures beyond academic research.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
3
|
Ebrahimi A, Ak G, Özel C, İzgördü H, Ghorbanpoor H, Hassan S, Avci H, Metintaş M. Clinical Perspectives and Novel Preclinical Models of Malignant Pleural Mesothelioma: A Critical Review. ACS Pharmacol Transl Sci 2024; 7:3299-3333. [PMID: 39539262 PMCID: PMC11555512 DOI: 10.1021/acsptsci.4c00324] [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: 05/31/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Pleural mesothelioma (PM), a rare malignant tumor explicitly associated with asbestos and erionite exposures, has become a global health problem due to limited treatment options and a poor prognosis, in which the median life expectancy varies depending on the method of treatment. However, the importance of early diagnosis is emphasized, and the practical methods have not matured yet. This study provides a critical overview of PM, addressing various aspects like epidemiology, etiology, diagnosis, treatment options, and the potential use of advanced technologies like microfluidic chip-based models for research and diagnosis. It initially begins with fundamentals of clinical aspects and then discusses the identification of disease-specific biomarkers in patients' serum or plasma samples, which could potentially be used for early diagnosis. A detailed investigation of the sophisticated preclinical models is highlighted. Recent three-dimensional (3D) model accomplishments, including microarchitecture modeling by transwell coculture, spheroids, organoids, 3D bioprinting constructs, and ex vivo tumor slices, are discussed comprehensively. On-chip models that imitate physiological processes, such as detection chips and therapeutic screening chips, are assessed as potential techniques. The review concludes with a critical and constructive discussion of the growing interest in the topic and its limitations and suggestions.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Güntülü Ak
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Ceren Özel
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Hüseyin İzgördü
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Hamed Ghorbanpoor
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Biomedical Engineering, Eskişehir
Osmangazi University, Eskişehir 26040, Turkey
| | - Shabir Hassan
- Department
of Biological Sciences, Khalifa University
of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Huseyin Avci
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Muzaffer Metintaş
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|
4
|
Thorel L, Divoux J, Lequesne J, Babin G, Morice PM, Florent R, Desmartin G, Lecouflet L, Marde Alagama C, Leconte A, Clarisse B, Briand M, Rouzier R, Gaichies L, Martin-Françoise S, Le Brun JF, Denoyelle C, Vigneron N, Jeanne C, Blanc-Fournier C, Leman R, Vaur D, Figeac M, Meryet-Figuiere M, Joly F, Weiswald LB, Poulain L, Dolivet E. The OVAREX study: Establishment of ex vivo ovarian cancer models to validate innovative therapies and to identify predictive biomarkers. BMC Cancer 2024; 24:701. [PMID: 38849726 PMCID: PMC11157894 DOI: 10.1186/s12885-024-12429-w] [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: 04/24/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Ovarian cancer is the first cause of death from gynecological malignancies mainly due to development of chemoresistance. Despite the emergence of PARP inhibitors, which have revolutionized the therapeutic management of some of these ovarian cancers, the 5-year overall survival rate remains around 45%. Therefore, it is crucial to develop new therapeutic strategies, to identify predictive biomarkers and to predict the response to treatments. In this context, functional assays based on patient-derived tumor models could constitute helpful and relevant tools for identifying efficient therapies or to guide clinical decision making. METHOD The OVAREX study is a single-center non-interventional study which aims at investigating the feasibility of establishing in vivo and ex vivo models and testing ex vivo models to predict clinical response of ovarian cancer patients. Patient-Derived Xenografts (PDX) will be established from tumor fragments engrafted subcutaneously into immunocompromised mice. Explants will be generated by slicing tumor tissues and Ascites-Derived Spheroids (ADS) will be isolated following filtration of ascites. Patient-derived tumor organoids (PDTO) will be established after dissociation of tumor tissues or ADS, cell embedding into extracellular matrix and culture in specific medium. Molecular and histological characterizations will be performed to compare tumor of origin and paired models. Response of ex vivo tumor-derived models to conventional chemotherapy and PARP inhibitors will be assessed and compared to results of companion diagnostic test and/or to the patient's response to evaluate their predictive value. DISCUSSION This clinical study aims at generating PDX and ex vivo models (PDTO, ADS, and explants) from tumors or ascites of ovarian cancer patients who will undergo surgical procedure or paracentesis. We aim at demonstrating the predictive value of ex vivo models for their potential use in routine clinical practice as part of precision medicine, as well as establishing a collection of relevant ovarian cancer models that will be useful for the evaluation of future innovative therapies. TRIAL REGISTRATION The clinical trial has been validated by local research ethic committee on January 25th 2019 and registered at ClinicalTrials.gov with the identifier NCT03831230 on January 28th 2019, last amendment v4 accepted on July 18, 2023.
Collapse
Affiliation(s)
- Lucie Thorel
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Jordane Divoux
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Justine Lequesne
- Clinical Research Department, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Guillaume Babin
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Department of Surgery, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Pierre-Marie Morice
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Romane Florent
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Guillaume Desmartin
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Lucie Lecouflet
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Chloé Marde Alagama
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
| | - Alexandra Leconte
- Clinical Research Department, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Bénédicte Clarisse
- Clinical Research Department, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Mélanie Briand
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Biological Resource Center 'OvaRessources', US PLATON, Université de Caen Normandie, Caen, France
| | - Roman Rouzier
- Department of Surgery, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Léopold Gaichies
- Department of Surgery, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | | | - Jean-François Le Brun
- Department of Surgery, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Christophe Denoyelle
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Nicolas Vigneron
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- Calvados General Tumor Registry, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Corinne Jeanne
- Department of Pathology, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Cécile Blanc-Fournier
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Biological Resource Center 'OvaRessources', US PLATON, Université de Caen Normandie, Caen, France
- Department of Pathology, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Raphaël Leman
- Department of Cancer Biology and Genetics, U1245 "Cancer and Brain Genomics", Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Dominique Vaur
- Department of Cancer Biology and Genetics, U1245 "Cancer and Brain Genomics", Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Martin Figeac
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Matthieu Meryet-Figuiere
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Florence Joly
- Clinical Research Department, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Louis-Bastien Weiswald
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France.
| | - Laurent Poulain
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- ORGAPRED Core Facility, US PLATON, Université de Caen Normandie, Caen, France.
- Biological Resource Center 'OvaRessources', US PLATON, Université de Caen Normandie, Caen, France.
| | - Enora Dolivet
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Department of Surgery, Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
| |
Collapse
|
5
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [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: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
6
|
Pecci V, Troisi F, Aiello A, De Martino S, Carlino A, Fiorentino V, Ripoli C, Rotili D, Pierconti F, Martini M, Porru M, Pinto F, Mai A, Bassi PF, Grassi C, Gaetano C, Pontecorvi A, Strigari L, Farsetti A, Nanni S. Targeting of H19/cell adhesion molecules circuitry by GSK-J4 epidrug inhibits metastatic progression in prostate cancer. Cancer Cell Int 2024; 24:56. [PMID: 38317193 PMCID: PMC10845766 DOI: 10.1186/s12935-024-03231-6] [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: 09/04/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND About 30% of Prostate cancer (PCa) patients progress to metastatic PCa that remains largely incurable. This evidence underlines the need for the development of innovative therapies. In this direction, the potential research focus might be on long non-coding RNAs (lncRNAs) like H19, which serve critical biological functions and show significant dysregulation in cancer. Previously, we showed a transcriptional down-regulation of H19 under combined pro-tumoral estrogen and hypoxia treatment in PCa cells that, in turn, induced both E-cadherin and β4 integrin expression. H19, indeed, acts as transcriptional repressor of cell adhesion molecules affecting the PCa metastatic properties. Here, we investigated the role of H19/cell adhesion molecules circuitry on in vivo PCa experimental tumor growth and metastatic dissemination models. METHODS H19 was silenced in luciferase-positive PC-3 and 22Rv1 cells and in vitro effect was evaluated by gene expression, proliferation and invasion assays before and after treatment with the histone lysine demethylase inhibitor, GSK-J4. In vivo tumor growth and metastasis dissemination, in the presence or absence of GSK-J4, were analyzed in two models of human tumor in immunodeficient mice by in vivo bioluminescent imaging and immunohistochemistry (IHC) on explanted tissues. Organotypic Slice Cultures (OSCs) from fresh PCa-explant were used as ex vivo model to test GSK-J4 effects. RESULTS H19 silencing in both PC-3 and 22Rv1 cells increased: i) E-cadherin and β4 integrin expression as well as proliferation and invasion, ii) in vivo tumor growth, and iii) metastasis formation at bone, lung, and liver. Of note, treatment with GSK-J4 reduced lesions. In parallel, GSK-J4 efficiently induced cell death in PCa-derived OSCs. CONCLUSIONS Our findings underscore the potential of the H19/cell adhesion molecules circuitry as a targeted approach in PCa treatment. Modulating this interaction has proven effective in inhibiting tumor growth and metastasis, presenting a logical foundation for targeted therapy.
Collapse
Affiliation(s)
- Valeria Pecci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy
| | - Fabiola Troisi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy
| | | | - Sara De Martino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy
- National Research Council (CNR)-IASI, Rome, Italy
| | - Angela Carlino
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
| | - Vincenzo Fiorentino
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
- Department of Woman, Child and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristian Ripoli
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Dante Rotili
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Francesco Pierconti
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
- Department of Woman, Child and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Martini
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
- Department of Woman, Child and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Manuela Porru
- Translational Oncology Research Unit, IRCCS- Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Pinto
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
| | - Antonello Mai
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Pier Francesco Bassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
| | - Claudio Grassi
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Alfredo Pontecorvi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy
| | - Lidia Strigari
- Department of Medical Physics, S. Orsola, Malpighi University Hospital, Bologna, Italy
| | | | - Simona Nanni
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, Rome, 00168, Italy.
- Fondazione "Policlinico Universitario A. Gemelli IRCCS", Rome, Italy.
| |
Collapse
|
7
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
8
|
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: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
9
|
Golan S, Bar V, Salpeter SJ, Neev G, Creiderman G, Kedar D, Aharon S, Turovsky L, Zundelevich A, Shahar H, Shapira H, Mallel G, Stossel E, Gavert N, Straussman R, Dotan Z, Berger R, Stossel C, Golan T, Halperin S, Leibovici D, Breuer S, Rottenberg Y, Applebaum L, Hubert A, Nechushtan H, Peretz T, Zick A, Chertin B, Koulikov D, Sonnenblick A, Rosenbaum E. A clinical evaluation of an ex vivo organ culture system to predict patient response to cancer therapy. Front Med (Lausanne) 2023; 10:1221484. [PMID: 37840996 PMCID: PMC10569691 DOI: 10.3389/fmed.2023.1221484] [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: 05/12/2023] [Accepted: 09/07/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Ex vivo organ cultures (EVOC) were recently optimized to sustain cancer tissue for 5 days with its complete microenvironment. We examined the ability of an EVOC platform to predict patient response to cancer therapy. Methods A multicenter, prospective, single-arm observational trial. Samples were obtained from patients with newly diagnosed bladder cancer who underwent transurethral resection of bladder tumor and from core needle biopsies of patients with metastatic cancer. The tumors were cut into 250 μM slices and cultured within 24 h, then incubated for 96 h with vehicle or intended to treat drug. The cultures were then fixed and stained to analyze their morphology and cell viability. Each EVOC was given a score based on cell viability, level of damage, and Ki67 proliferation, and the scores were correlated with the patients' clinical response assessed by pathology or Response Evaluation Criteria in Solid Tumors (RECIST). Results The cancer tissue and microenvironment, including endothelial and immune cells, were preserved at high viability with continued cell division for 5 days, demonstrating active cell signaling dynamics. A total of 34 cancer samples were tested by the platform and were correlated with clinical results. A higher EVOC score was correlated with better clinical response. The EVOC system showed a predictive specificity of 77.7% (7/9, 95% CI 0.4-0.97) and a sensitivity of 96% (24/25, 95% CI 0.80-0.99). Conclusion EVOC cultured for 5 days showed high sensitivity and specificity for predicting clinical response to therapy among patients with muscle-invasive bladder cancer and other solid tumors.
Collapse
Affiliation(s)
- Shay Golan
- Department of Urology, Beilinson Hospital – Rabin Medical Center, Petah Tikva, Israel
| | | | | | | | - German Creiderman
- Department of Urology, Beilinson Hospital – Rabin Medical Center, Petah Tikva, Israel
| | - Daniel Kedar
- Department of Urology, Beilinson Hospital – Rabin Medical Center, Petah Tikva, Israel
| | | | | | | | | | | | | | | | - Nancy Gavert
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ravid Straussman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Zohar Dotan
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Raanan Berger
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Chani Stossel
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Talia Golan
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Sharon Halperin
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Dan Leibovici
- Department of Urology, Kaplan Medical Center, Rehovot, Israel
| | - Shani Breuer
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yakir Rottenberg
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liat Applebaum
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ayala Hubert
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hovav Nechushtan
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamar Peretz
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aviad Zick
- Sharett Institute of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Boris Chertin
- Department of Urology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Dmitry Koulikov
- Department of Urology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Amir Sonnenblick
- Department of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
10
|
Huang YL, Dickerson LK, Kenerson H, Jiang X, Pillarisetty V, Tian Q, Hood L, Gujral TS, Yeung RS. Organotypic Models for Functional Drug Testing of Human Cancers. BME FRONTIERS 2023; 4:0022. [PMID: 37849667 PMCID: PMC10275620 DOI: 10.34133/bmef.0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/30/2023] [Indexed: 10/19/2023] Open
Abstract
In the era of personalized oncology, there have been accelerated efforts to develop clinically relevant platforms to test drug sensitivities of individual cancers. An ideal assay will serve as a diagnostic companion to inform the oncologist of the various treatments that are sensitive and insensitive, thus improving outcome while minimizing unnecessary toxicities and costs. To date, no such platform exists for clinical use, but promising approaches are on the horizon that take advantage of improved techniques in creating human cancer models that encompass the entire tumor microenvironment, alongside technologies for assessing and analyzing tumor response. This review summarizes a number of current strategies that make use of intact human cancer tissues as organotypic cultures in drug sensitivity testing.
Collapse
Affiliation(s)
- Yu Ling Huang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Heidi Kenerson
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Xiuyun Jiang
- Department of Surgery, University of Washington, Seattle, WA, USA
| | | | - Qiang Tian
- National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leroy Hood
- Institute for Systems Biology, Phenome Health Institute, Seattle, WA, USA
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Dong M, Böpple K, Thiel J, Winkler B, Liang C, Schueler J, Davies EJ, Barry ST, Metsalu T, Mürdter TE, Sauer G, Ott G, Schwab M, Aulitzky WE. Perfusion Air Culture of Precision-Cut Tumor Slices: An Ex Vivo System to Evaluate Individual Drug Response under Controlled Culture Conditions. Cells 2023; 12:cells12050807. [PMID: 36899943 PMCID: PMC10001200 DOI: 10.3390/cells12050807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Precision-cut tumor slices (PCTS) maintain tissue heterogeneity concerning different cell types and preserve the tumor microenvironment (TME). Typically, PCTS are cultured statically on a filter support at an air-liquid interface, which gives rise to intra-slice gradients during culture. To overcome this problem, we developed a perfusion air culture (PAC) system that can provide a continuous and controlled oxygen medium, and drug supply. This makes it an adaptable ex vivo system for evaluating drug responses in a tissue-specific microenvironment. PCTS from mouse xenografts (MCF-7, H1437) and primary human ovarian tumors (primary OV) cultured in the PAC system maintained the morphology, proliferation, and TME for more than 7 days, and no intra-slice gradients were observed. Cultured PCTS were analyzed for DNA damage, apoptosis, and transcriptional biomarkers for the cellular stress response. For the primary OV slices, cisplatin treatment induced a diverse increase in the cleavage of caspase-3 and PD-L1 expression, indicating a heterogeneous response to drug treatment between patients. Immune cells were preserved throughout the culturing period, indicating that immune therapy can be analyzed. The novel PAC system is suitable for assessing individual drug responses and can thus be used as a preclinical model to predict in vivo therapy responses.
Collapse
Affiliation(s)
- Meng Dong
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
- Correspondence: ; Tel.: +49-711-8101-2070
| | - Kathrin Böpple
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
| | - Julia Thiel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
| | - Bernd Winkler
- Department of Gynecology and Obstetrics, Robert Bosch Hospital, 70376 Stuttgart, Germany
| | - Chunguang Liang
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Julia Schueler
- Charles River Germany GmbH, Am Flughafen 12-14, 79108 Freiburg, Germany
| | - Emma J. Davies
- Bioscience, Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK
| | - Simon T. Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK
| | - Tauno Metsalu
- Institute of Computer Science, University of Tartu, 51009 Tartu, Estonia
| | - Thomas E. Mürdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
| | - Georg Sauer
- Department of Gynecology and Obstetrics, Robert Bosch Hospital, 70376 Stuttgart, Germany
| | - German Ott
- Department of Clinical Pathology, Robert Bosch Hospital, 70376 Stuttgart, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
- Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | | |
Collapse
|
12
|
Jungwirth G, Hanemann CO, Dunn IF, Herold-Mende C. Preclinical Models of Meningioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1416:199-211. [PMID: 37432629 DOI: 10.1007/978-3-031-29750-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The management of clinically aggressive meningiomas remains challenging due to limited treatment options aside from surgical removal and radiotherapy. High recurrence rates and lack of effective systemic therapies contribute to the unfavorable prognosis of these patients. Accurate in vitro and in vivo models are critical for understanding meningioma pathogenesis and to identify and test novel therapeutics. In this chapter, we review cell models, genetically engineered mouse models, and xenograft mouse models, with special emphasis on the field of application. Finally, promising preclinical 3D models such as organotypic tumor slices and patient-derived tumor organoids are discussed.
Collapse
Affiliation(s)
- Gerhard Jungwirth
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany.
| | - C Oliver Hanemann
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, UK
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
13
|
Advances in application and innovation of microfluidic platforms for pharmaceutical analysis. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
14
|
Role of Patient-Derived Models of Cancer in Translational Oncology. Cancers (Basel) 2022; 15:cancers15010139. [PMID: 36612135 PMCID: PMC9817860 DOI: 10.3390/cancers15010139] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/04/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer is a heterogeneous disease. Each individual tumor is unique and characterized by structural, cellular, genetic and molecular features. Therefore, patient-derived cancer models are indispensable tools in cancer research and have been actively introduced into the healthcare system. For instance, patient-derived models provide a good reproducibility of susceptibility and resistance of cancer cells against drugs, allowing personalized therapy for patients. In this article, we review the advantages and disadvantages of the following patient-derived models of cancer: (1) PDC-patient-derived cell culture, (2) PDS-patient-derived spheroids and PDO-patient-derived organoids, (3) PDTSC-patient-derived tissue slice cultures, (4) PDX-patient-derived xenografts, humanized PDX, as well as PDXC-PDX-derived cell cultures and PDXO-PDX-derived organoids. We also provide an overview of current clinical investigations and new developments in the area of patient-derived cancer models. Moreover, attention is paid to databases of patient-derived cancer models, which are collected in specialized repositories. We believe that the widespread use of patient-derived cancer models will improve our knowledge in cancer cell biology and contribute to the development of more effective personalized cancer treatment strategies.
Collapse
|
15
|
An oncogene addiction phosphorylation signature and its derived scores inform tumor responsiveness to targeted therapies. Cell Mol Life Sci 2022; 80:6. [PMID: 36494469 PMCID: PMC9734221 DOI: 10.1007/s00018-022-04634-2] [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: 02/18/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Oncogene addiction provides important therapeutic opportunities for precision oncology treatment strategies. To date the cellular circuitries associated with driving oncoproteins, which eventually establish the phenotypic manifestation of oncogene addiction, remain largely unexplored. Data suggest the DNA damage response (DDR) as a central signaling network that intersects with pathways associated with deregulated addicting oncoproteins with kinase activity in cancer cells. EXPERIMENTAL DESIGN: We employed a targeted mass spectrometry approach to systematically explore alterations in 116 phosphosites related to oncogene signaling and its intersection with the DDR following inhibition of the addicting oncogene alone or in combination with irradiation in MET-, EGFR-, ALK- or BRAF (V600)-positive cancer models. An NSCLC tissue pipeline combining patient-derived xenografts (PDXs) and ex vivo patient organotypic cultures has been established for treatment responsiveness assessment. RESULTS We identified an 'oncogene addiction phosphorylation signature' (OAPS) consisting of 8 protein phosphorylations (ACLY S455, IF4B S422, IF4G1 S1231, LIMA1 S490, MYCN S62, NCBP1 S22, P3C2A S259 and TERF2 S365) that are significantly suppressed upon targeted oncogene inhibition solely in addicted cell line models and patient tissues. We show that the OAPS is present in patient tissues and the OAPS-derived score strongly correlates with the ex vivo responses to targeted treatments. CONCLUSIONS We propose a score derived from OAPS as a quantitative measure to evaluate oncogene addiction of cancer cell samples. This work underlines the importance of protein phosphorylation assessment for patient stratification in precision oncology and corresponding identification of tumor subtypes sensitive to inhibition of a particular oncogene.
Collapse
|
16
|
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
|
17
|
Blessing WA, Digesu CS, Liu R, Mahvi DA, Tal-Mason A, Kumar A, Hachey KJ, Colby AH, Korunes-Miller JT, Agar N, Regan MS, Shih A, Raut CP, Grinstaff MW, Colson YL. Sustained Supratherapeutic Paclitaxel Delivery Enhances Irreversible Sarcoma Cell Death. Mol Cancer Ther 2022; 21:1663-1673. [PMID: 36031342 PMCID: PMC9633561 DOI: 10.1158/1535-7163.mct-21-0750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 05/26/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022]
Abstract
Risk of locoregional recurrence after sarcoma resection is high, increasing both morbidity and mortality. Intraoperative implantation of paclitaxel (PTX)-eluting polymer films locally delivers sustained, supratherapeutic PTX concentrations to the tumor bed that are not clinically feasible with systemic therapy, thereby reducing recurrence and improving survival in a murine model of recurrent sarcoma. However, the biology underlying increased efficacy of PTX-eluting films is unknown and provides the impetus for this work. In vitro PTX efficacy is time and dose dependent with prolonged exposure significantly decreasing PTX IC50 values for human chondrosarcoma (CS-1) cells (153.9 nmol/L at 4 hours vs. 14.2 nmol/L at 30 hours, P = 0.0001). High-dose PTX significantly inhibits proliferation with in vivo PTX films delivering a dose >130 μmol/L directly to the tumor thereby irreversibly arresting cell cycle and inducing apoptosis in CS-1 as well as patient-derived liposarcoma (LP6) and leiomyosarcoma (LMS20). Supratherapeutic PTX upregulates the expression of p21 in G2-M arrested cells, and irreversibly induces apoptosis followed by cell death, within 4 hours of exposure. Microarray analyses corroborate the finding of poor DNA integrity commonly observed as a final step of apoptosis in CS-1 cells and tumor. Unlike low PTX concentrations at the tumor bed during systemic delivery, supratherapeutic concentrations achieved with PTX-eluting films markedly decrease sarcoma lethality in vivo and offer an alternative paradigm to prevent recurrence.
Collapse
Affiliation(s)
- William A. Blessing
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Christopher S. Digesu
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Rong Liu
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - David A. Mahvi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Aya Tal-Mason
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Anil Kumar
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Aaron H. Colby
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA
| | - Jenny T. Korunes-Miller
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA
| | - Natalie Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Michael S. Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Angela Shih
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Chandrajit P. Raut
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Perréard M, Florent R, Thorel L, Vincent A, Weiswald LB, Poulain L. Les organoïdes dérivés de tumeurs (ou tumoroïdes), des outils de choix pour la médecine de précision en oncologie. Med Sci (Paris) 2022; 38:888-895. [DOI: 10.1051/medsci/2022149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Il est désormais possible d’établir des tumoroïdes à partir de presque tout type de tumeur, notamment en vue de la mise en place de tests fonctionnels prédictifs et/ou de l’identification de signatures moléculaires prédictives. Bien que l’optimisation des conditions de culture ou la complexification du micro-environnement des tumoroïdes soit encore nécessaire, de nombreuses applications sont déjà envisageables dans le domaine de la prédiction de la réponse aux traitements et de l’orientation de la décision thérapeutique. Par l’introduction de leur utilisation en clinique, l’oncologie de précision pourrait bien entrer dans une nouvelle ère dans le courant de la décennie à venir.
Collapse
|
19
|
Weitz J, Hurtado de Mendoza T, Tiriac H, Lee J, Sun S, Garg B, Patel J, Li K, Baumgartner J, Kelly KJ, Veerapong J, Hosseini M, Chen Y, Lowy AM. An Ex Vivo Organotypic Culture Platform for Functional Interrogation of Human Appendiceal Cancer Reveals a Prominent and Heterogenous Immunological Landscape. Clin Cancer Res 2022; 28:4793-4806. [PMID: 36067351 DOI: 10.1158/1078-0432.ccr-22-0980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/04/2022] [Accepted: 08/31/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Epithelial neoplasms of the appendix are difficult to study preclinically given their low incidence, frequent mucinous histology, and absence of a comparable organ in mice for disease modeling. Although surgery is an effective treatment for localized disease, metastatic disease has a poor prognosis as existing therapeutics borrowed from colorectal cancer have limited efficacy. Recent studies reveal that appendiceal cancer has a genomic landscape distinct from colorectal cancer and thus preclinical models to study this disease are a significant unmet need. EXPERIMENTAL DESIGN We adopted an ex vivo slice model that permits the study of cellular interactions within the tumor microenvironment. Mucinous carcinomatosis peritonei specimens obtained at surgical resection were cutoff using a vibratome to make 150-μm slices cultured in media. RESULTS Slice cultures were viable and maintained their cellular composition regarding the proportion of epithelial, immune cells, and fibroblasts over 7 days. Within donor specimens, we identified a prominent and diverse immune landscape and calcium imaging confirmed that immune cells were functional for 7 days. Given the diverse immune landscape, we treated slices with TAK981, an inhibitor of SUMOylation with known immunomodulatory functions, in early-phase clinical trials. In 5 of 6 donor samples, TAK981-treated slices cultures had reduced viability, and regulatory T cells (Treg). These data were consistent with TAK981 activity in purified Tregs using an in vitro murine model. CONCLUSIONS This study demonstrates an approach to study appendiceal cancer therapeutics and pathobiology in a preclinical setting. These methods may be broadly applicable to the study of other malignancies.
Collapse
Affiliation(s)
- Jonathan Weitz
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Tatiana Hurtado de Mendoza
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Herve Tiriac
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - James Lee
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Siming Sun
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Bharti Garg
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Jay Patel
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Kevin Li
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Joel Baumgartner
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Kaitlin J Kelly
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Jula Veerapong
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Mojgan Hosseini
- Department of Pathology and Laboratory Medicine, University of California, San Diego, California
| | - Yuan Chen
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| | - Andrew M Lowy
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, California
| |
Collapse
|
20
|
Tsolova AO, Aguilar RM, Maybin JA, Critchley HOD. Pre-clinical models to study abnormal uterine bleeding (AUB). EBioMedicine 2022; 84:104238. [PMID: 36081283 PMCID: PMC9465267 DOI: 10.1016/j.ebiom.2022.104238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
Abnormal Uterine Bleeding (AUB) is a common debilitating condition that significantly reduces quality of life of women across the reproductive age span. AUB creates significant morbidity, medical, social, and economic problems for women, their families, workplace, and health services. Despite the profoundly negative effects of AUB on public health, advancement in understanding the pathophysiology of AUB and the discovery of novel effective therapies is slow due to lack of reliable pre-clinical models. This review discusses currently available laboratory-based pre-clinical scientific models and how they are used to study AUB. Human and animal in vitro, ex vivo, and in vivo models will be described along with advantages and limitations of each method.
Collapse
|
21
|
Jubelin C, Muñoz-Garcia J, Griscom L, Cochonneau D, Ollivier E, Heymann MF, Vallette FM, Oliver L, Heymann D. Three-dimensional in vitro culture models in oncology research. Cell Biosci 2022; 12:155. [PMID: 36089610 PMCID: PMC9465969 DOI: 10.1186/s13578-022-00887-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer is a multifactorial disease that is responsible for 10 million deaths per year. The intra- and inter-heterogeneity of malignant tumors make it difficult to develop single targeted approaches. Similarly, their diversity requires various models to investigate the mechanisms involved in cancer initiation, progression, drug resistance and recurrence. Of the in vitro cell-based models, monolayer adherent (also known as 2D culture) cell cultures have been used for the longest time. However, it appears that they are often less appropriate than the three-dimensional (3D) cell culture approach for mimicking the biological behavior of tumor cells, in particular the mechanisms leading to therapeutic escape and drug resistance. Multicellular tumor spheroids are widely used to study cancers in 3D, and can be generated by a multiplicity of techniques, such as liquid-based and scaffold-based 3D cultures, microfluidics and bioprinting. Organoids are more complex 3D models than multicellular tumor spheroids because they are generated from stem cells isolated from patients and are considered as powerful tools to reproduce the disease development in vitro. The present review provides an overview of the various 3D culture models that have been set up to study cancer development and drug response. The advantages of 3D models compared to 2D cell cultures, the limitations, and the fields of application of these models and their techniques of production are also discussed.
Collapse
|
22
|
Disulfiram increases the efficacy of 5-fluorouracil in organotypic cultures of colorectal carcinoma. Biomed Pharmacother 2022; 153:113465. [DOI: 10.1016/j.biopha.2022.113465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022] Open
|
23
|
Moro CF, Selvam AK, Ghaderi M, Pimenoff VN, Gerling M, Bozóky B, Elduayen SP, Dillner J, Björnstedt M. Drug-induced tumor-specific cytotoxicity in a whole tissue ex vivo model of human pancreatic ductal adenocarcinoma. Front Oncol 2022; 12:965182. [PMID: 36059619 PMCID: PMC9436406 DOI: 10.3389/fonc.2022.965182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. PDAC has a dismal prognosis and an inherent resistance to cytostatic drugs. The lack of reliable experimental models is a severe limitation for drug development targeting PDAC. We have employed a whole tissue ex vivo culture model to explore the effect of redox-modulation by sodium selenite on the viability and growth of PDAC. Drug-resistant tumors are more vulnerable to redox-active selenium compounds because of high metabolic activity and redox imbalance. Sodium selenite efficiently and specifically reduced PDAC cell viability (p <0.02) (n=8) and decreased viable de novo tumor cell outgrowth (p<0.05) while preserving non-neoplastic tissues. Major cellular responses (damaged tumor cells > 90%, tumor regression grades III-IV according to Evans) were observed for sodium selenite concentrations between 15-30 µM. Moreover, selenium levels used in this study were significantly below the previously reported maximum tolerated dose for humans. Transcriptome data analysis revealed decreased expression of genes known to drive PDAC growth and metastatic potential (CEMIP, DDR2, PLOD2, P4HA1) while the cell death-inducing genes (ATF3, ACHE) were significantly upregulated (p<0.0001). In conclusion, we report that sodium selenite has an extraordinary efficacy and specificity against drug-resistant pancreatic cancer in an organotypic slice culture model. Our ex vivo organotypic tissue slice culture model can be used to test a variety of drug candidates for swift and reliable drug responses to individual PDAC cases.
Collapse
Affiliation(s)
- Carlos Fernández Moro
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Arun Kumar Selvam
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mehran Ghaderi
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Ville N. Pimenoff
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Gerling
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Tema Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Béla Bozóky
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Soledad Pouso Elduayen
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Joakim Dillner
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Björnstedt
- Department of Laboratory Medicine, Division of Pathology F46, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
- *Correspondence: Mikael Björnstedt,
| |
Collapse
|
24
|
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
|
25
|
Preclinical models of epithelial ovarian cancer: practical considerations and challenges for a meaningful application. Cell Mol Life Sci 2022; 79:364. [PMID: 35705879 PMCID: PMC9200670 DOI: 10.1007/s00018-022-04395-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/05/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022]
Abstract
Despite many improvements in ovarian cancer diagnosis and treatment, until now, conventional chemotherapy and new biological drugs have not been shown to cure the disease, and the overall prognosis remains poor. Over 90% of ovarian malignancies are categorized as epithelial ovarian cancers (EOC), a collection of different types of neoplasms with distinctive disease biology, response to chemotherapy, and outcome. Advances in our understanding of the histopathology and molecular features of EOC subtypes, as well as the cellular origins of these cancers, have given a boost to the development of clinically relevant experimental models. The overall goal of this review is to provide a comprehensive description of the available preclinical investigational approaches aimed at better characterizing disease development and progression and at identifying new therapeutic strategies. Systems discussed comprise monolayer (2D) and three-dimensional (3D) cultures of established and primary cancer cell lines, organoids and patient-derived explants, animal models, including carcinogen-induced, syngeneic, genetically engineered mouse, xenografts, patient-derived xenografts (PDX), humanized PDX, and the zebrafish and the laying hen models. Recent advances in tumour-on-a-chip platforms are also detailed. The critical analysis of strengths and weaknesses of each experimental model will aid in identifying opportunities to optimize their translational value.
Collapse
|
26
|
Three-dimensional (3D) liver cell models - a tool for bridging the gap between animal studies and clinical trials when screening liver accumulation and toxicity of nanobiomaterials. Drug Deliv Transl Res 2022; 12:2048-2074. [PMID: 35507131 PMCID: PMC9066991 DOI: 10.1007/s13346-022-01147-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/13/2022]
Abstract
Despite the exciting properties and wide-reaching applications of nanobiomaterials (NBMs) in human health and medicine, their translation from bench to bedside is slow, with a predominant issue being liver accumulation and toxicity following systemic administration. In vitro 2D cell-based assays and in vivo testing are the most popular and widely used methods for assessing liver toxicity at pre-clinical stages; however, these fall short in predicting toxicity for NBMs. Focusing on in vitro and in vivo assessment, the accurate prediction of human-specific hepatotoxicity is still a significant challenge to researchers. This review describes the relationship between NBMs and the liver, and the methods for assessing toxicity, focusing on the limitations they bring in the assessment of NBM hepatotoxicity as one of the reasons defining the poor translation for NBMs. We will then present some of the most recent advances towards the development of more biologically relevant in vitro liver methods based on tissue-mimetic 3D cell models and how these could facilitate the translation of NBMs going forward. Finally, we also discuss the low public acceptance and limited uptake of tissue-mimetic 3D models in pre-clinical assessment, despite the demonstrated technical and ethical advantages associated with them.
Collapse
|
27
|
Parsian M, Mutlu P, Yildirim E, Ildiz C, Ozen C, Gunduz U. Development of a microfluidic platform to maintain viability of micro-dissected tumor slices in culture. BIOMICROFLUIDICS 2022; 16:034103. [PMID: 35547184 PMCID: PMC9076128 DOI: 10.1063/5.0087532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/06/2022] [Indexed: 05/07/2023]
Abstract
One of the issues limiting the development of personalized medicine is the absence of realistic models that reflect the nature and complexity of tumor tissues. We described a new tissue culture approach that combines a microfluidic chip with the microdissected breast cancer tumor. "Tumor-on-a-chip" devices are suitable for precision medicine since the viability of tissue samples is maintained during the culture period by continuously feeding fresh media and eliminating metabolic wastes from the tissue. However, the mass transport of oxygen, which arguably is the most critical nutrient, is rarely assessed. According to our results, transportation of oxygen provides satisfactory in vivo oxygenation within the system. A high level of dissolved oxygen, around 98%-100% for every 24 h, was measurable in the outlet medium. The microfluidic chip system developed within the scope of this study allows living and testing tumor tissues under laboratory conditions. In this study, tumors were generated in CD-1 mice using MDA-MB-231 and SKBR-3 cell lines. Microdissected tumor tissues were cultured both in the newly developed microfluidic chip system and in conventional 24-well culture plates. Two systems were compared for two different types of tumors. The confocal microscopy analyses, lactate dehydrogenase release, and glucose consumption values showed that the tissues in the microfluidic system remained more viable with respect to the conventional well plate culturing method, up to 96 h. The new culturing technique described here may be superior to conventional culturing techniques for developing new treatment strategies, such as testing chemotherapeutics on tumor samples from individual patients.
Collapse
Affiliation(s)
- Maryam Parsian
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | - Pelin Mutlu
- Department of Biotechnology, Ankara University, Ankara, Turkey
- Author to whom correspondence should be addressed:
| | - Ender Yildirim
- Department of Mechanical Engineering, Middle East Technical University, Ankara, Turkey
| | - Can Ildiz
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Can Ozen
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | | |
Collapse
|
28
|
Precision-Cut Tumor Slices (PCTS) as an Ex Vivo Model in Immunotherapy Research. Antibodies (Basel) 2022; 11:antib11020026. [PMID: 35466279 PMCID: PMC9036232 DOI: 10.3390/antib11020026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/04/2022] Open
Abstract
Precision-cut tumor slices (PCTS) have recently emerged as important ex vivo human tumor models, offering the opportunity to study individual patient responses to targeted immunotherapies, including CAR-T cell therapies. In this review, an outline of different human tumor models available in laboratory settings is provided, with a focus on the unique characteristics of PCTS. Standard PCTS generation and maintenance procedures are outlined, followed by an in-depth overview of PCTS utilization in preclinical research aiming to better understand the unique functional characteristics of cytotoxic T cells within human tumors. Furthermore, recent studies using PCTS as an ex vivo model for predicting patient responses to immunotherapies and other targeted therapies against solid tumors are thoroughly presented. Finally, the advantages and limitations of the PCTS models are discussed. PCTS are expected to gain momentum and be fully utilized as a significant tool towards better patient stratification and personalized medicine.
Collapse
|
29
|
Xue C, Corey E, Gujral TS. Proteomic and Transcriptomic Profiling Reveals Mitochondrial Oxidative Phosphorylation as Therapeutic Vulnerability in Androgen Receptor Pathway Active Prostate Tumors. Cancers (Basel) 2022; 14:cancers14071739. [PMID: 35406510 PMCID: PMC8997167 DOI: 10.3390/cancers14071739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Metastatic prostate cancer (PC) is the second leading cause of cancer deaths in males. The lack of preclinical models and molecular characterization for advanced stage PC is a key barrier in understanding the aggressive subsets androgen receptor (AR) pathway active or AR-null castration-resistant prostate cancers (CRPC). Our study aimed to assess the potential of patient-derived xenograft (PDX) models and an approach integrating proteomic and transcriptomic techniques to explore the underlying drivers of metastatic PC. Transcriptomic and proteomic profiling of 42 PDX prostate tumors uncovered both previously established and unexpected molecular features of aggressive PC subsets. Of these, we confirmed the functional role of mitochondrial metabolism in AR-positive CRPC. Abstract Metastatic prostate cancer (PC) is the second leading cause of cancer deaths in males and has limited therapeutic options. The lack of preclinical models for advanced stage PC represents one of the primary barriers in understanding the key genetic drivers of aggressive subsets, including androgen receptor (AR) pathway active and AR-null castration-resistant prostate cancers (CRPC). In our studies, we described a series of LuCaP patient-derived xenograft (PDX) models representing the major genomic and phenotypic features of human disease. To fully exploit the potential of these preclinical models, we carried out a comprehensive transcriptomic and proteomic profiling of 42 LuCaP PDX prostate tumors. The collected proteomic data (~6000 data points) based on 71 antibodies revealed many of the previously known molecular markers associated with AR-positive and AR-null CRPC. Genomic analysis indicated subtype-specific activation of pathways such as Wnt/beta-catenin signaling, mTOR, and oxidative phosphorylation for AR-positive CRPC and upregulation of carbohydrate metabolism and glucose metabolism for AR-null CRPC. Of these, we functionally confirmed the role of mitochondrial metabolism in AR-positive CRPC cell lines. Our data highlight how the integration of transcriptomic and proteomic approaches and PDX systems as preclinical models can potentially map the connectivity of poorly understood signaling pathways in metastatic prostate cancer.
Collapse
Affiliation(s)
- Caroline Xue
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA;
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Correspondence:
| |
Collapse
|
30
|
Zhang Y, Wang ZY, Jing HS, Zhang HD, Yan HX, Fan JX, Zhai B. A pre‑clinical model combining cryopreservation technique with precision‑cut slice culture method to assess the in vitro drug response of hepatocellular carcinoma. Int J Mol Med 2022; 49:51. [PMID: 35179217 PMCID: PMC8904079 DOI: 10.3892/ijmm.2022.5107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
Models considering hepatocellular carcinoma (HCC) complexity cannot be accurately replicated in routine cell lines or animal models. We aimed to evaluate the practicality of tissue slice culture by combining it with a cryopreservation technique. We prepared 0.3-mm-thick tissue slices by a microtome and maintained their cell viability using a cryopreservation technique. Slices were cultured individually in the presence or absence of regorafenib (REG) for 72 h. Alterations in morphology and gene expression were assessed by histological and genetic analysis. Overall viability was also analyzed in tissue slices by CCK-8 quantification assay and fluorescent staining. Tissue morphology and cell viability were evaluated to quantify drug effects. Histological and genetic analyses showed that no significant alterations in morphology and gene expression were induced by the vitrification-based cryopreservation method. The viability of warmed HCC tissues was up to 90% of the fresh tissues. The viability and proliferation could be retained for at least four days in the filter culture system. The positive drug responses in precision-cut slice culture in vitro were evaluated by tissue morphology and cell viability. In summary, the successful application of precision-cut HCC slice culture combined with a cryopreservation technique in a systematic drug screening demonstrates the feasibility and utility of slice culture method for assessing drug response.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Zhen-Yu Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Shu Jing
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Dan Zhang
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai 200120, P.R. China
| | - He-Xin Yan
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian-Xia Fan
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Bo Zhai
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
31
|
Xu J, Wu PJ, Lai TH, Sharma P, Canella A, Welker AM, Beattie C, Timmers CD, Lang FF, Jacob NK, Elder JB, Lonser R, Easley M, Pietrzak M, Sampath D, Puduvalli VK. Disruption of DNA Repair and Survival Pathways through Heat Shock Protein inhibition by Onalespib to Sensitize Malignant Gliomas to Chemoradiation therapy. Clin Cancer Res 2022; 28:1979-1990. [PMID: 35140124 PMCID: PMC9064967 DOI: 10.1158/1078-0432.ccr-20-0468] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/10/2021] [Accepted: 02/04/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Proficient DNA repair by homologous recombination (HR) facilitates resistance to chemo-radiation in glioma stem cells (GSCs). We evaluated whether compromising HR by targeting HSP90, a molecular chaperone required for the function of key HR proteins, using onalespib, a long-acting, brain-penetrant HSP90 inhibitor, would sensitize high-grade gliomas to chemo-radiation in vitro and in vivo Experimental Design: The ability of onalespib to deplete HR client proteins, impair HR repair capacity, and sensitize GBM to chemo-radiation was evaluated in vitro in GSCs, and in vivo using zebrafish and mouse intracranial glioma xenograft models. The effects of HSP90 inhibition on the transcriptome and cytoplasmic proteins was assessed in GSCs and in ex vivo organotypic human glioma slice cultures. RESULTS Treatment with onalespib depleted CHK1 and RAD51, two key proteins of the HR pathway, and attenuated HR repair, sensitizing GSCs to the combination of radiation and temozolomide (TMZ). HSP90 inhibition reprogrammed the transcriptome of GSCs and broadly altered expression of cytoplasmic proteins including known and novel client proteins relevant to GSCs. The combination of onalespib with radiation and TMZ extended survival in a zebra fish and a mouse xenograft model of GBM compared to the standard of care (radiation and TMZ) or onalespib with radiation. CONCLUSIONS The results of this study demonstrate that targeting HR by HSP90 inhibition sensitizes GSCs to radiation and chemotherapy and extends survival in zebrafish and mouse intracranial models of GBM. These results provide a preclinical rationale for assessment of HSP90 inhibitors in combination with chemoradiation in GBM patients.
Collapse
Affiliation(s)
- Jihong Xu
- Neuro-Oncology, The University of Texas MD Anderson Cancer Center
| | - Pei-Jung Wu
- Division of Neuro-oncology, The Ohio State University
| | - Tzung-Huei Lai
- Division of Hematology, Department of Medicine, The Ohio State University
| | - Pratibha Sharma
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center
| | | | | | | | | | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center
| | - Naduparambil K Jacob
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center
| | - J Bradley Elder
- Dardinger Neuro-Oncology Center, Department of Neurosurgery, The Ohio State University
| | - Russell Lonser
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke
| | | | | | - Deepa Sampath
- Hematopoeitic Biology and Malignancy, The University of Texas MD Anderson Cancer Center
| | - Vinay K Puduvalli
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
32
|
Functional Therapeutic Target Validation Using Pediatric Zebrafish Xenograft Models. Cancers (Basel) 2022; 14:cancers14030849. [PMID: 35159116 PMCID: PMC8834194 DOI: 10.3390/cancers14030849] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite the major progress of precision and personalized oncology, a significant therapeutic benefit is only achieved in cases with directly druggable genetic alterations. This highlights the need for additional methods that reliably predict each individual patient’s response in a clinically meaningful time, e.g., through ex vivo functional drug screen profiling. Moreover, patient-derived xenograft (PDX) models are more predictive than cell culture studies, as they reconstruct cell–cell and cell–extracellular matrix (ECM) interactions and consider the tumor microenvironment, drug metabolism and toxicities. Zebrafish PDXs (zPDX) are nowadays emerging as a fast model allowing for multiple drugs to be tested at the same time in a clinically relevant time window. Here, we show that functional drug response profiling of zPDX from primary material obtained through the INdividualized Therapy FOr Relapsed Malignancies in Childhood (INFORM) pediatric precision oncology pipeline provides additional key information for personalized precision oncology. Abstract The survival rate among children with relapsed tumors remains poor, due to tumor heterogeneity, lack of directly actionable tumor drivers and multidrug resistance. Novel personalized medicine approaches tailored to each tumor are urgently needed to improve cancer treatment. Current pediatric precision oncology platforms, such as the INFORM (INdividualized Therapy FOr Relapsed Malignancies in Childhood) study, reveal that molecular profiling of tumor tissue identifies targets associated with clinical benefit in a subgroup of patients only and should be complemented with functional drug testing. In such an approach, patient-derived tumor cells are exposed to a library of approved oncological drugs in a physiological setting, e.g., in the form of animal avatars injected with patient tumor cells. We used molecularly fully characterized tumor samples from the INFORM study to compare drug screen results of individual patient-derived cell models in functional assays: (i) patient-derived spheroid cultures within a few days after tumor dissociation; (ii) tumor cells reisolated from the corresponding mouse PDX; (iii) corresponding long-term organoid-like cultures and (iv) drug evaluation with the corresponding zebrafish PDX (zPDX) model. Each model had its advantage and complemented the others for drug hit and drug combination selection. Our results provide evidence that in vivo zPDX drug screening is a promising add-on to current functional drug screening in precision medicine platforms.
Collapse
|
33
|
Ex vivo organotypic cultures for synergistic therapy prioritization identify patient-specific responses to combined MEK and Src inhibition in colorectal cancer. NATURE CANCER 2022; 3:219-231. [PMID: 35145327 DOI: 10.1038/s43018-021-00325-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022]
Abstract
Translating preclinical studies to effective treatment protocols and identifying specific therapeutic responses in individuals with cancer is challenging. This may arise due to the complex genetic makeup of tumor cells and the impact of their multifaceted tumor microenvironment on drug response. To find new clinically relevant drug combinations for colorectal cancer (CRC), we prioritized the top five synergistic combinations from a large in vitro screen for ex vivo testing on 29 freshly resected human CRC tumors and found that only the combination of mitogen-activated protein kinase kinase (MEK) and proto-oncogene tyrosine-protein kinase Src (Src) inhibition was effective when tested ex vivo. Pretreatment phosphorylated Src (pSrc) was identified as a predictive biomarker for MEK and Src inhibition only in the absence of KRASG12 mutations. Overall, we demonstrate the potential of using ex vivo platforms to identify drug combinations and discover MEK and Src dual inhibition as an effective drug combination in a predefined subset of individuals with CRC.
Collapse
|
34
|
Claridge SE, Cavallo JA, Hopkins BD. Patient-Derived In Vitro and In Vivo Models of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:215-233. [DOI: 10.1007/978-3-030-91836-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
De Ridder K, Tung N, Werle JT, Karpf L, Awad RM, Bernier A, Ceuppens H, Salmon H, Goyvaerts C. Novel 3D Lung Tumor Spheroids for Oncoimmunological Assays. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy Department of Biomedical Sciences Vrije Universiteit Brussel Laarbeeklaan 103-E 1090 Jette Belgium
| | - Navpreet Tung
- Department of Oncological Sciences The Precision Immunology Institute The Tisch Cancer Institute Icahn School of Medicine at Mount Sinai 1470 Madison Avenue New York NY 10029 USA
| | - Jan-Timon Werle
- Institut Curie INSERM 75005 Paris France
- PSL Research University 75006 Paris France
| | - Léa Karpf
- Department of Oncological Sciences The Precision Immunology Institute The Tisch Cancer Institute Icahn School of Medicine at Mount Sinai 1470 Madison Avenue New York NY 10029 USA
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy Department of Biomedical Sciences Vrije Universiteit Brussel Laarbeeklaan 103-E 1090 Jette Belgium
| | - Annie Bernier
- Institut Curie INSERM 75005 Paris France
- PSL Research University 75006 Paris France
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy Department of Biomedical Sciences Vrije Universiteit Brussel Laarbeeklaan 103-E 1090 Jette Belgium
| | - Hélène Salmon
- Department of Oncological Sciences The Precision Immunology Institute The Tisch Cancer Institute Icahn School of Medicine at Mount Sinai 1470 Madison Avenue New York NY 10029 USA
- Institut Curie INSERM 75005 Paris France
- PSL Research University 75006 Paris France
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy Department of Biomedical Sciences Vrije Universiteit Brussel Laarbeeklaan 103-E 1090 Jette Belgium
| |
Collapse
|
36
|
Varrone F, Mandrich L, Caputo E. Melanoma Immunotherapy and Precision Medicine in the Era of Tumor Micro-Tissue Engineering: Where Are We Now and Where Are We Going? Cancers (Basel) 2021; 13:5788. [PMID: 34830940 PMCID: PMC8616100 DOI: 10.3390/cancers13225788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma still remains a cancer with very poor survival rates, although it is at the forefront of personalized medicine. Most patients show partial responses and disease progressed due to adaptative resistance mechanisms, preventing long-lasting clinical benefits to the current treatments. The response to therapies can be shaped by not only taking into account cancer cell heterogeneity and plasticity, but also by its structural context as well as the cellular component of the tumor microenvironment (TME). Here, we review the recent development in the field of immunotherapy and target-based therapy and how, in the era of tumor micro-tissue engineering, ex-vivo assays could help to enhance our melanoma biology knowledge in its complexity, translating it in the development of successful therapeutic strategies, as well as in the prediction of therapeutic benefits.
Collapse
Affiliation(s)
| | - Luigi Mandrich
- Research Institute on Terrestrial Ecosystem—IRET-CNR Via Pietro Castellino 111, I-80131 Naples, Italy;
| | - Emilia Caputo
- Institute of Genetics and Biophysics—IGB-CNR, “A. Buzzati-Traverso”, Via Pietro Castellino 111, I-80131 Naples, Italy
| |
Collapse
|
37
|
Romualdo GR, Leroy K, Costa CJS, Prata GB, Vanderborght B, da Silva TC, Barbisan LF, Andraus W, Devisscher L, Câmara NOS, Vinken M, Cogliati B. In Vivo and In Vitro Models of Hepatocellular Carcinoma: Current Strategies for Translational Modeling. Cancers (Basel) 2021; 13:5583. [PMID: 34771745 PMCID: PMC8582701 DOI: 10.3390/cancers13215583] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer-related death globally. HCC is a complex multistep disease and usually emerges in the setting of chronic liver diseases. The molecular pathogenesis of HCC varies according to the etiology, mainly caused by chronic hepatitis B and C virus infections, chronic alcohol consumption, aflatoxin-contaminated food, and non-alcoholic fatty liver disease associated with metabolic syndrome or diabetes mellitus. The establishment of HCC models has become essential for both basic and translational research to improve our understanding of the pathophysiology and unravel new molecular drivers of this disease. The ideal model should recapitulate key events observed during hepatocarcinogenesis and HCC progression in view of establishing effective diagnostic and therapeutic strategies to be translated into clinical practice. Despite considerable efforts currently devoted to liver cancer research, only a few anti-HCC drugs are available, and patient prognosis and survival are still poor. The present paper provides a state-of-the-art overview of in vivo and in vitro models used for translational modeling of HCC with a specific focus on their key molecular hallmarks.
Collapse
Affiliation(s)
- Guilherme Ribeiro Romualdo
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Kaat Leroy
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Cícero Júlio Silva Costa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Gabriel Bacil Prata
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
| | - Wellington Andraus
- Department of Gastroenterology, Clinics Hospital, School of Medicine, University of São Paulo (HC-FMUSP), São Paulo 05403-000, Brazil;
| | - Lindsey Devisscher
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| |
Collapse
|
38
|
Aguilar Cosme JR, Gagui DC, Green NH, Bryant HE, Claeyssens F. In Vitro Low-Fluence Photodynamic Therapy Parameter Screening Using 3D Tumor Spheroids Shows that Fractionated Light Treatments Enhance Phototoxicity. ACS Biomater Sci Eng 2021; 7:5078-5089. [PMID: 34615346 DOI: 10.1021/acsbiomaterials.1c00690] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The evaluation of novel photosensitizers (PSs) for photodynamic therapy (PDT) is difficult due to the limitations of two-dimensional cell culture and multiple parameters (dose, light intensity, uptake time), which complicate progression to in vivo experiments and clinical translation. Three-dimensional (3D) cell culture models like multicellular cancer tumor spheroids (MCTS) show great similarities to in vivo avascular tumor conditions, improving the speed and accuracy of screening novel compounds with various treatment combinations. In this study, we utilize C8161 human melanoma spheroids to screen PDT treatment combinations using protoporphyrin IX (PpIX) and drug-loaded carbon dot (CD) conjugates PpIX-CD and PpIX@CD at ultralow fluence values (<10 J/cm2). Conjugates show equivalent light-induced damage to PpIX from 1 μg/mL with significantly less dark cytotoxicity up to 72 h after exposure, shown by LDH release and dsDNA content. Fractionated treatments, carried out by dividing light exposure with 24 h intervals, demonstrate an enhanced PDT effect compared to single exposure at equal concentrations. Light sheet fluorescence microscopy combined with live/dead staining demonstrates that spheroids sustain extensive damage after PDT, with PpIX and PpIX-CD showing improved uptake compared to PpIX@CD. We show that PDT parameter screening can be carried out using a low-cost and convenient combination of assays to improve the efficiency of evaluating novel compounds.
Collapse
Affiliation(s)
- Jose R Aguilar Cosme
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Dan C Gagui
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Nicola H Green
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Helen E Bryant
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom
| | - Frederik Claeyssens
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
39
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
40
|
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
|
41
|
Using ex vivo culture to assess dynamic phenotype changes in human prostate macrophages following exposure to therapeutic drugs. Sci Rep 2021; 11:19299. [PMID: 34588590 PMCID: PMC8481239 DOI: 10.1038/s41598-021-98903-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 09/09/2021] [Indexed: 01/01/2023] Open
Abstract
Within the prostate tumor microenvironment (TME) there are complex multi-faceted and dynamic communication occurring between cancer cells and immune cells. Macrophages are key cells which infiltrate and surround tumor cells and are recognized to significantly contribute to tumor resistance and metastases. Our understanding of their function in the TME is commonly based on in vitro and in vivo models, with limited research to confirm these model observations in human prostates. Macrophage infiltration was evaluated within the TME of human prostates after 72 h culture of fresh biopsies samples in the presence of control or enzalutamide. In addition to immunohistochemistry, an optimized protocol for multi-parametric evaluation of cellular surface markers was developed using flow cytometry. Flow cytometry parameters were compared to clinicopathological features. Immunohistochemistry staining for 19 patients with paired samples suggested enzalutamide increased the expression of CD163 relative to CD68 staining. Techniques to validate these results using flow cytometry of dissociated biopsies after 72 h of culture are described. In a second cohort of patients with Gleason grade group ≥ 3 prostate cancer, global macrophage expression of CD163 was unchanged with enzalutamide treatment. However, exploratory analyses of our results using multi-parametric flow cytometry for multiple immunosuppressive macrophage markers suggest subgroup changes as well as novel associations between circulating biomarkers like the neutrophil to lymphocyte ratio (NLR) and immune cell phenotype composition in the prostate TME. Further, we observed an association between B7–H3 expressing tumor-associated macrophages and the presence of intraductal carcinoma. The use of flow cytometry to evaluate ex vivo cultured prostate biopsies fills an important gap in our ability to understand the immune cell composition of the prostate TME. Our results highlight novel associations for further investigation.
Collapse
|
42
|
Ando Y, Mariano C, Shen K. Engineered in vitro tumor models for cell-based immunotherapy. Acta Biomater 2021; 132:345-359. [PMID: 33857692 PMCID: PMC8434941 DOI: 10.1016/j.actbio.2021.03.076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022]
Abstract
Tumor immunotherapy is rapidly evolving as one of the major pillars of cancer treatment. Cell-based immunotherapies, which utilize patient's own immune cells to eliminate cancer cells, have shown great promise in treating a range of malignancies, especially those of hematopoietic origins. However, their performance on a broader spectrum of solid tumor types still fall short of expectations in the clinical stage despite promising preclinical assessments. In this review, we briefly introduce cell-based immunotherapies and the inhibitory mechanisms in tumor microenvironments that may have contributed to this discrepancy. Specifically, a major obstacle to the clinical translation of cell-based immunotherapies is in the lack of preclinical models that can accurately assess the efficacies and mechanisms of these therapies in a (patho-)physiologically relevant manner. Lately, tissue engineering and organ-on-a-chip tools and microphysiological models have allowed for more faithful recapitulation of the tumor microenvironments, by incorporating crucial tumor tissue features such as cellular phenotypes, tissue architecture, extracellular matrix, physical parameters, and their dynamic interactions. This review summarizes the existing engineered tumor models with a focus on tumor immunology and cell-based immunotherapy. We also discuss some key considerations for the future development of engineered tumor models for immunotherapeutics. STATEMENT OF SIGNIFICANCE: Cell-based immunotherapies have shown great promise in treating hematological malignancies and some epithelial tumors. However, their performance on a broader spectrum of solid tumor types still fall short of expectations. Major obstacles include the inhibitory mechanisms in tumor microenvironments (TME) and the lack of preclinical models that can accurately assess the efficacies and mechanisms of cellular therapies in a (patho-)physiologically relevant manner. In this review, we introduce recent progress in tissue engineering and microphysiological models for more faithful recapitulation of TME for cell-based immunotherapies, and some key considerations for the future development of engineered tumor models. This overview will provide a better understanding on the role of engineered models in accelerating immunotherapeutic discoveries and clinical translations.
Collapse
Affiliation(s)
- Yuta Ando
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Chelsea Mariano
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; USC Stem Cell, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
43
|
Suckert T, Nexhipi S, Dietrich A, Koch R, Kunz-Schughart LA, Bahn E, Beyreuther E. Models for Translational Proton Radiobiology-From Bench to Bedside and Back. Cancers (Basel) 2021; 13:4216. [PMID: 34439370 PMCID: PMC8395028 DOI: 10.3390/cancers13164216] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022] Open
Abstract
The number of proton therapy centers worldwide are increasing steadily, with more than two million cancer patients treated so far. Despite this development, pending questions on proton radiobiology still call for basic and translational preclinical research. Open issues are the on-going discussion on an energy-dependent varying proton RBE (relative biological effectiveness), a better characterization of normal tissue side effects and combination treatments with drugs originally developed for photon therapy. At the same time, novel possibilities arise, such as radioimmunotherapy, and new proton therapy schemata, such as FLASH irradiation and proton mini-beams. The study of those aspects demands for radiobiological models at different stages along the translational chain, allowing the investigation of mechanisms from the molecular level to whole organisms. Focusing on the challenges and specifics of proton research, this review summarizes the different available models, ranging from in vitro systems to animal studies of increasing complexity as well as complementing in silico approaches.
Collapse
Affiliation(s)
- Theresa Suckert
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sindi Nexhipi
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01309 Dresden, Germany
| | - Antje Dietrich
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Robin Koch
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Emanuel Bahn
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, 69120 Heidelberg, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiation Physics, 01328 Dresden, Germany
| |
Collapse
|
44
|
Dorrigiv D, Simeone K, Communal L, Kendall-Dupont J, St-Georges-Robillard A, Péant B, Carmona E, Mes-Masson AM, Gervais T. Microdissected Tissue vs Tissue Slices-A Comparative Study of Tumor Explant Models Cultured On-Chip and Off-Chip. Cancers (Basel) 2021; 13:4208. [PMID: 34439362 PMCID: PMC8394960 DOI: 10.3390/cancers13164208] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/29/2022] Open
Abstract
Predicting patient responses to anticancer drugs is a major challenge both at the drug development stage and during cancer treatment. Tumor explant culture platforms (TECPs) preserve the native tissue architecture and are well-suited for drug response assays. However, tissue longevity in these models is relatively low. Several methodologies have been developed to address this issue, although no study has compared their efficacy in a controlled fashion. We investigated the effect of two variables in TECPs, specifically, the tissue size and culture vessel on tissue survival using micro-dissected tumor tissue (MDT) and tissue slices which were cultured in microfluidic chips and plastic well plates. Tumor models were produced from ovarian and prostate cancer cell line xenografts and were matched in terms of the specimen, total volume of tissue, and respective volume of medium in each culture system. We examined morphology, viability, and hypoxia in the various tumor models. Our observations suggest that the viability and proliferative capacity of MDTs were not affected during the time course of the experiments. In contrast, tissue slices had reduced proliferation and showed increased cell death and hypoxia under both culture conditions. Tissue slices cultured in microfluidic devices had a lower degree of hypoxia compared to those in 96-well plates. Globally, our results show that tissue slices have lower survival rates compared to MDTs due to inherent diffusion limitations, and that microfluidic devices may decrease hypoxia in tumor models.
Collapse
Affiliation(s)
- Dina Dorrigiv
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Institute of Biomedical Engineering Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | - Kayla Simeone
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Laudine Communal
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Jennifer Kendall-Dupont
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Amélie St-Georges-Robillard
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | - Benjamin Péant
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Euridice Carmona
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Thomas Gervais
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Institute of Biomedical Engineering Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
45
|
Liu D, Hou X, Wu W, Zanfagnin V, Li Y, Correia C, Zhao Z, Zhao C, Liu Z, Zhang T, Fang Z, Wang H, Xu C, Weroha SJ, Kaufmann SH, Dai H. Constitutive BAK/MCL1 complexes predict paclitaxel and S63845 sensitivity of ovarian cancer. Cell Death Dis 2021; 12:789. [PMID: 34385422 PMCID: PMC8361168 DOI: 10.1038/s41419-021-04073-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/08/2021] [Accepted: 04/26/2021] [Indexed: 12/03/2022]
Abstract
We previously found that preformed complexes of BAK with antiapoptotic BCL2 proteins predict BH3 mimetic sensitivities in lymphohematopoietic cells. These complexes have not previously been examined in solid tumors or in the context of conventional anticancer drugs. Here we show the relative amount of BAK found in preformed complexes with MCL1 or BCLXL varies across ovarian cancer cell lines and patient-derived xenografts (PDXs). Cells bearing BAK/MCL1 complexes were more sensitive to paclitaxel and the MCL1 antagonist S63845. Likewise, PDX models with BAK/MCL1 complexes were more likely to respond to paclitaxel. Mechanistically, BIM induced by low paclitaxel concentrations interacted preferentially with MCL1 and displaced MCL1-bound BAK. Further studies indicated that cells with preformed BAK/MCL1 complexes were sensitive to the paclitaxel/S63845 combination, while cells without BAK/MCL1 complexes were not. Our study suggested that the assessment of BAK/MCL1 complexes might be useful for predicting response to paclitaxel alone or in combination with BH3 mimetics.
Collapse
Affiliation(s)
- Dongyan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wangyu Wu
- Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | | | - Yunjian Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zhiyang Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chenggang Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Zhirong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Tao Zhang
- Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhiyou Fang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chao Xu
- University of Science and Technology of China, Hefei, 230026, China
| | - Saravut J Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Haiming Dai
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
46
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
47
|
Zhao W, Dovas A, Spinazzi EF, Levitin HM, Banu MA, Upadhyayula P, Sudhakar T, Marie T, Otten ML, Sisti MB, Bruce JN, Canoll P, Sims PA. Deconvolution of cell type-specific drug responses in human tumor tissue with single-cell RNA-seq. Genome Med 2021; 13:82. [PMID: 33975634 PMCID: PMC8114529 DOI: 10.1186/s13073-021-00894-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Preclinical studies require models that recapitulate the cellular diversity of human tumors and provide insight into the drug sensitivities of specific cellular populations. The ideal platform would enable rapid screening of cell type-specific drug sensitivities directly in patient tumor tissue and reveal strategies to overcome intratumoral heterogeneity. METHODS We combine multiplexed drug perturbation in acute slice culture from freshly resected tumors with single-cell RNA sequencing (scRNA-seq) to profile transcriptome-wide drug responses in individual patients. We applied this approach to drug perturbations on slices derived from six glioblastoma (GBM) resections to identify conserved drug responses and to one additional GBM resection to identify patient-specific responses. RESULTS We used scRNA-seq to demonstrate that acute slice cultures recapitulate the cellular and molecular features of the originating tumor tissue and the feasibility of drug screening from an individual tumor. Detailed investigation of etoposide, a topoisomerase poison, and the histone deacetylase (HDAC) inhibitor panobinostat in acute slice cultures revealed cell type-specific responses across multiple patients. Etoposide has a conserved impact on proliferating tumor cells, while panobinostat treatment affects both tumor and non-tumor populations, including unexpected effects on the immune microenvironment. CONCLUSIONS Acute slice cultures recapitulate the major cellular and molecular features of GBM at the single-cell level. In combination with scRNA-seq, this approach enables cell type-specific analysis of sensitivity to multiple drugs in individual tumors. We anticipate that this approach will facilitate pre-clinical studies that identify effective therapies for solid tumors.
Collapse
Affiliation(s)
- Wenting Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Athanassios Dovas
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Hanna Mendes Levitin
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matei Alexandru Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Pavan Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Tejaswi Sudhakar
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Tamara Marie
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Marc L Otten
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Michael B Sisti
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Peter Canoll
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
48
|
Sim J, Lee HJ, Jeong B, Park MH. Poly(Ethylene Glycol)-Poly(l-Alanine)/Hyaluronic Acid Complex as a 3D Platform for Understanding Cancer Cell Migration in the Tumor Microenvironment. Polymers (Basel) 2021; 13:polym13071042. [PMID: 33810521 PMCID: PMC8036448 DOI: 10.3390/polym13071042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/01/2023] Open
Abstract
Cancer progression and migration in the tumor microenvironment are related to cell types and three-dimensional (3D) matrices. Therefore, developing biomimetic tumor models, including co-culture systems and a tunable 3D matrix, could play an essential role in understanding the cancer environment. Here, multicellular spheroids using human adipose-derived mesenchymal stem cells (hADSCs) and breast cancer cells (MDA-MB-231) within the 3D matrix were used as a tumor microenvironment (TME) mimicking platform. The amphiphilic peptide block copolymer and hyaluronic acid (HA) formed a self-assembled structure, which provides a biocompatible 3D environment for the cells. Multicellular spheroids were formed on the optimized plate and were observed as cell migration from a spheroid within a 3D matrix, such as the invasive and metastatic cancer of TME. This study suggests a new 3D platform using polymer complexes and the importance of tumor complexities, including various cell types and microenvironments.
Collapse
Affiliation(s)
- Jooyoung Sim
- Center for Convergence Bioceramic Materials, Korea Institute of Ceramic Engineering and Technology, Cheongju 28160, Korea;
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea;
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea;
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea;
- Correspondence: (B.J.); (M.H.P.); Tel.: +82-2-3277-3411 (B.J.); +82-43-913-1515 (M.H.P.)
| | - Min Hee Park
- Center for Convergence Bioceramic Materials, Korea Institute of Ceramic Engineering and Technology, Cheongju 28160, Korea;
- Correspondence: (B.J.); (M.H.P.); Tel.: +82-2-3277-3411 (B.J.); +82-43-913-1515 (M.H.P.)
| |
Collapse
|
49
|
Advani D, Sharma S, Kumari S, Ambasta RK, Kumar P. Precision Oncology, Signaling and Anticancer Agents in Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:433-468. [PMID: 33687887 DOI: 10.2174/1871520621666210308101029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The global alliance for genomics and healthcare facilities provides innovational solutions to expedite research and clinical practices for complex and incurable health conditions. Precision oncology is an emerging field explicitly tailored to facilitate cancer diagnosis, prevention and treatment based on patients' genetic profile. Advancements in "omics" techniques, next-generation sequencing, artificial intelligence and clinical trial designs provide a platform for assessing the efficacy and safety of combination therapies and diagnostic procedures. METHOD Data were collected from Pubmed and Google scholar using keywords: "Precision medicine", "precision medicine and cancer", "anticancer agents in precision medicine" and reviewed comprehensively. RESULTS Personalized therapeutics including immunotherapy, cancer vaccines, serve as a groundbreaking solution for cancer treatment. Herein, we take a measurable view of precision therapies and novel diagnostic approaches targeting cancer treatment. The contemporary applications of precision medicine have also been described along with various hurdles identified in the successful establishment of precision therapeutics. CONCLUSION This review highlights the key breakthroughs related to immunotherapies, targeted anticancer agents, and target interventions related to cancer signaling mechanisms. The success story of this field in context to drug resistance, safety, patient survival and in improving quality of life is yet to be elucidated. We conclude that, in the near future, the field of individualized treatments may truly revolutionize the nature of cancer patient care.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| |
Collapse
|
50
|
Cooperative Blockade of CK2 and ATM Kinases Drives Apoptosis in VHL-Deficient Renal Carcinoma Cells through ROS Overproduction. Cancers (Basel) 2021; 13:cancers13030576. [PMID: 33540838 PMCID: PMC7867364 DOI: 10.3390/cancers13030576] [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: 01/14/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) is the eighth leading malignancy in the world, accounting for 4% of all cancers with poor outcome when metastatic. Protein kinases are highly druggable proteins, which are often aberrantly activated in cancers. The aim of our study was to identify candidate targets for metastatic clear cell renal cell carcinoma therapy, using chemo-genomic-based high-throughput screening. We found that the combined inhibition of the CK2 and ATM kinases in renal tumor cells and patient-derived tumor samples induces synthetic lethality. Mechanistic investigations unveil that this drug combination triggers apoptosis through HIF-2α-(Hypoxic inducible factor HIF-2α) dependent reactive oxygen species (ROS) overproduction, giving a new option for patient care in metastatic RCC. Abstract Kinase-targeted agents demonstrate antitumor activity in advanced metastatic clear cell renal cell carcinoma (ccRCC), which remains largely incurable. Integration of genomic approaches through small-molecules and genetically based high-throughput screening holds the promise of improved discovery of candidate targets for cancer therapy. The 786-O cell line represents a model for most ccRCC that have a loss of functional pVHL (von Hippel-Lindau). A multiplexed assay was used to study the cellular fitness of a panel of engineered ccRCC isogenic 786-O VHL− cell lines in response to a collection of targeted cancer therapeutics including kinase inhibitors, allowing the interrogation of over 2880 drug–gene pairs. Among diverse patterns of drug sensitivities, investigation of the mechanistic effect of one selected drug combination on tumor spheroids and ex vivo renal tumor slice cultures showed that VHL-defective ccRCC cells were more vulnerable to the combined inhibition of the CK2 and ATM kinases than wild-type VHL cells. Importantly, we found that HIF-2α acts as a key mediator that potentiates the response to combined CK2/ATM inhibition by triggering ROS-dependent apoptosis. Importantly, our findings reveal a selective killing of VHL-deficient renal carcinoma cells and provide a rationale for a mechanism-based use of combined CK2/ATM inhibitors for improved patient care in metastatic VHL-ccRCC.
Collapse
|