1
|
Zuo J, Fang Y, Wang R, Liang S. High-throughput solutions in tumor organoids: from culture to drug screening. Stem Cells 2025; 43:sxae070. [PMID: 39460616 PMCID: PMC11811636 DOI: 10.1093/stmcls/sxae070] [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: 07/19/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Tumor organoids have emerged as an ideal in vitro model for patient-derived tissues, as they recapitulate the characteristics of the source tumor tissue to a certain extent, offering the potential for personalized tumor therapy and demonstrating significant promise in pharmaceutical research and development. However, establishing and applying this model involves multiple labor-intensive and time-consuming experimental steps and lacks standardized protocols and uniform identification criteria. Thus, high-throughput solutions are essential for the widespread adoption of tumor organoid models. This review provides a comprehensive overview of current high-throughput solutions across the entire workflow of tumor organoids, from sampling and culture to drug screening. Furthermore, we explore various technologies that can control and optimize single-cell preparation, organoid culture, and drug screening with the ultimate goal of ensuring the automation and high efficiency of the culture system and identifying more effective tumor therapeutics.
Collapse
Affiliation(s)
- Jianing Zuo
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| | - Yanhua Fang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Ruoyu Wang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| | - Shanshan Liang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| |
Collapse
|
2
|
Li Y, Wu W, Yao J, Wang S, Wu X, Yan J. Patient-Derived Tumor Organoids: A Platform for Precision Therapy of Colorectal Cancer. Cell Transplant 2025; 34:9636897251314645. [PMID: 39953837 PMCID: PMC11829288 DOI: 10.1177/09636897251314645] [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: 10/31/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 02/17/2025] Open
Abstract
Colorectal cancer (CRC) represents a significant cause of cancer-related mortality on a global scale. It is a highly heterogeneous cancer, and the response of patients to homogeneous drug therapy varies considerably. Patient-derived tumor organoids (PDTOs) represent an optimal preclinical model for cancer research. A substantial body of evidence from numerous studies has demonstrated that PDTOs can accurately predict a patient's response to different drug treatments. This article outlines the utilization of PDTOs in the management of CRC across a range of therapeutic contexts, including postoperative adjuvant chemotherapy, palliative chemotherapy, neoadjuvant chemoradiotherapy, targeted therapy, third-line and follow-up treatment, and the treatment of elderly patients. This article delineates the manner in which PDTOs can inform therapeutic decisions at all stages of CRC, thereby assisting clinicians in selecting treatment options and reducing the risk of toxicity and resistance associated with clinical drugs. Moreover, it identifies shortcomings of existing PDTOs, including the absence of consistent criteria for assessing drug sensitivity tests, the lack of vascular and tumor microenvironment models, and the high cost of the technology. In conclusion, despite their inherent limitations, PDTOs offer several advantages, including rapid culture, a high success rate, high consistency, and high throughput, which can be employed as a personalized treatment option for CRC. The use of PDTOs in CRC allows for the prediction of responses to different treatment modalities at various stages of disease progression. This has the potential to reduce adverse drug reactions and the emergence of resistance associated with clinical drugs, facilitate evidence-based clinical decision-making, and guide CRC patients in the selection of personalized medications, thereby advancing the individualized treatment of CRC.
Collapse
Affiliation(s)
- Yiran Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Wei Wu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jiaxin Yao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Suidong Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiufeng Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, P.R. China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| |
Collapse
|
3
|
Löwenmark T, Köhn L, Kellgren T, Rosenbaum W, Bronnec V, Löfgren-Burström A, Zingmark C, Larsson P, Dahlberg M, Schroeder BO, Wai SN, Ljuslinder I, Edin S, Palmqvist R. Parvimonas micra forms a distinct bacterial network with oral pathobionts in colorectal cancer patients. J Transl Med 2024; 22:947. [PMID: 39420333 PMCID: PMC11487773 DOI: 10.1186/s12967-024-05720-8] [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: 07/02/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Mounting evidence suggests a significant role of the gut microbiota in the development and progression of colorectal cancer (CRC). In particular, an over-representation of oral pathogens has been linked to CRC. The aim of this study was to further investigate the faecal microbial landscape of CRC patients, with a focus on the oral pathogens Parvimonas micra and Fusobacterium nucleatum. METHODS In this study, 16S rRNA sequencing was conducted using faecal samples from CRC patients (n = 275) and controls without pathological findings (n = 95). RESULTS We discovered a significant difference in microbial composition depending on tumour location and microsatellite instability (MSI) status, with P. micra, F. nucleatum, and Peptostreptococcus stomatis found to be more abundant in patients with MSI tumours. Moreover, P. micra and F. nucleatum were associated with a cluster of CRC-related bacteria including Bacteroides fragilis as well as with other oral pathogens such as P. stomatis and various Porphyromonas species. This cluster was distinctly different in the control group, suggesting its potential linkage with CRC. CONCLUSIONS Our results suggest a similar distribution of several CRC-associated bacteria within CRC patients, underscoring the importance of considering the concomitant presence of bacterial species in studies investigating the mechanisms of CRC development and progression.
Collapse
Affiliation(s)
- Thyra Löwenmark
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Linda Köhn
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Therese Kellgren
- Department of Mathematics and Mathematical Statistics, Umeå University, Umeå, Sweden
| | - William Rosenbaum
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Vicky Bronnec
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pär Larsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Michael Dahlberg
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | | | - Sun Nyunt Wai
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Ingrid Ljuslinder
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden.
| |
Collapse
|
4
|
van der Graaff D, Seghers S, Vanclooster P, Deben C, Vandamme T, Prenen H. Advancements in Research and Treatment Applications of Patient-Derived Tumor Organoids in Colorectal Cancer. Cancers (Basel) 2024; 16:2671. [PMID: 39123399 PMCID: PMC11311786 DOI: 10.3390/cancers16152671] [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: 07/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant health burden globally, being the second leading cause of cancer-related mortality. Despite significant therapeutic advancements, resistance to systemic antineoplastic agents remains an important obstacle, highlighting the need for innovative screening tools to tailor patient-specific treatment. This review explores the application of patient-derived tumor organoids (PDTOs), three-dimensional, self-organizing models derived from patient tumor samples, as screening tools for drug resistance in CRC. PDTOs offer unique advantages over traditional models by recapitulating the tumor architecture, cellular heterogeneity, and genomic landscape and are a valuable ex vivo predictive drug screening tool. This review provides an overview of the current literature surrounding the use of PDTOs as an instrument for predicting therapy responses in CRC. We also explore more complex models, such as co-cultures with important stromal cells, such as cancer-associated fibroblasts, and organ-on-a-chip models. Furthermore, we discuss the use of PDTOs for drug repurposing, offering a new approach to identify the existing drugs effective against drug-resistant CRC. Additionally, we explore how PDTOs serve as models to gain insights into drug resistance mechanisms, using newer techniques, such as single-cell RNA sequencing and CRISPR-Cas9 genome editing. Through this review, we aim to highlight the potential of PDTOs in advancing our understanding of predicting therapy responses, drug resistance, and biomarker identification in CRC management.
Collapse
Affiliation(s)
| | - Sofie Seghers
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | | | - Christophe Deben
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | - Timon Vandamme
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | - Hans Prenen
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
5
|
Liu Y, Wang D, Luan Y, Tao B, Li Q, Feng Q, Zhou H, Mu J, Yu J. The application of organoids in colorectal diseases. Front Pharmacol 2024; 15:1412489. [PMID: 38983913 PMCID: PMC11231380 DOI: 10.3389/fphar.2024.1412489] [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: 04/05/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Intestinal organoids are a three-dimensional cell culture model derived from colon or pluripotent stem cells. Intestinal organoids constructed in vitro strongly mimic the colon epithelium in cell composition, tissue architecture, and specific functions, replicating the colon epithelium in an in vitro culture environment. As an emerging biomedical technology, organoid technology has unique advantages over traditional two-dimensional culture in preserving parental gene expression and mutation, cell function, and biological characteristics. It has shown great potential in the research and treatment of colorectal diseases. Organoid technology has been widely applied in research on colorectal topics, including intestinal tumors, inflammatory bowel disease, infectious diarrhea, and intestinal injury regeneration. This review focuses on the application of organoid technology in colorectal diseases, including the basic principles and preparation methods of organoids, and explores the pathogenesis of and personalized treatment plans for various colorectal diseases to provide a valuable reference for organoid technology development and application.
Collapse
Affiliation(s)
- Yanxin Liu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Yanhong Luan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Boqiang Tao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Jianfeng Mu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Jinhai Yu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
HUANG Z, LI B, WANG Y, XUE J, WEI Z, LIANG N, LI S. [Application and Research Progress of Lung Cancer Organoid in Precision Medicine
for Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:276-282. [PMID: 38769830 PMCID: PMC11110296 DOI: 10.3779/j.issn.1009-3419.2024.106.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Indexed: 05/22/2024]
Abstract
The continuous advancement of molecular detection technology has greatly propelled the development of precision medicine for lung cancer. However, tumor heterogeneity is closely associated with tumor metastasis, recurrence, and drug resistance. Additionally, different lung cancer patients with the same genetic mutation may exhibit varying treatment responses to different therapeutic strategies. Therefore, the development of modern precision medicine urgently requires the precise formulation of personalized treatment strategies through personalized tumor models. Lung cancer organoid (LCO) can highly simulate the biological characteristics of tumor in vivo, facilitating the application of innovative drugs such as antibody-drug conjugate in precision medicine for lung cancer. With the development of co-culture model of LCO with tumor microenvironment and tissue engineering technology such as microfluidic chip, LCO can better preserve the biological characteristics and functions of tumor tissue, further improving high-throughput and automated drug sensitivity experiment. In this review, we combine the latest research progress to summarize the application progress and challenges of LCO in precision medicine for lung cancer.
.
Collapse
|
7
|
Guo L, Li C, Gong W. Toward reproducible tumor organoid culture: focusing on primary liver cancer. Front Immunol 2024; 15:1290504. [PMID: 38571961 PMCID: PMC10987700 DOI: 10.3389/fimmu.2024.1290504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Organoids present substantial potential for pushing forward preclinical research and personalized medicine by accurately recapitulating tissue and tumor heterogeneity in vitro. However, the lack of standardized protocols for cancer organoid culture has hindered reproducibility. This paper comprehensively reviews the current challenges associated with cancer organoid culture and highlights recent multidisciplinary advancements in the field with a specific focus on standardizing liver cancer organoid culture. We discuss the non-standardized aspects, including tissue sources, processing techniques, medium formulations, and matrix materials, that contribute to technical variability. Furthermore, we emphasize the need to establish reproducible platforms that accurately preserve the genetic, proteomic, morphological, and pharmacotypic features of the parent tumor. At the end of each section, our focus shifts to organoid culture standardization in primary liver cancer. By addressing these challenges, we can enhance the reproducibility and clinical translation of cancer organoid systems, enabling their potential applications in precision medicine, drug screening, and preclinical research.
Collapse
Affiliation(s)
| | | | - Weiqiang Gong
- Department of Hepatobiliary and Pancreatic Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
8
|
Chen H, Yang Y, Shi J, Yan T, Wang J, Yang Y, Lu Q, Feng H, Du J, Cao Z, Weygant N. Comparison of Surgical and Colonoscopy Tissue to Establish Colorectal Patient-derived Organoids. Curr Cancer Drug Targets 2024; 24:546-555. [PMID: 37997804 DOI: 10.2174/0115680096263866231024112120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/25/2023] [Accepted: 09/13/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Patient-derived organoids (PDOs) are ex vivo models that retain the functions and characteristics of individualized source tissues, including a simulated tumor microenvironment. However, the potential impact of undiscovered differences between tissue sources on PDO growth and progression remains unclear. OBJECTIVE This study aimed to compare the growth and condition of PDO models originating from surgical resection and colonoscopy and to provide practical insights for PDO studies. METHODS Tissue samples and relevant patient clinical information were collected to establish organoid models. PDOs were derived from both surgical and colonoscopy tissues. The growth of the organoids, including their state, size, and success rate of establishment, was recorded and analyzed. The activity of the organoids at the end stage of growth was detected using calcein-AM fluorescence staining. RESULTS The results showed that the early growth phase of 2/3 colonoscopy-derived organoids was faster compared to surgical PDOs, with a growth difference observed within 11-13 days of establishment. However, colonoscopy-derived organoids exhibited a diminished growth trend after this time. There were no significant differences observed in the terminal area and quantity between the two types of tissue-derived organoids. Immunofluorescence assays of the PDOs revealed that the surgical PDOs possessed a denser cell mass with relatively higher viability than colonoscopy-derived PDOs. CONCLUSION In the establishment of colorectal patient-derived organoids, surgically derived organoids require a slightly longer establishment period, while colonoscopy-derived organoids should be passaged prior to growth inhibition to preserve organoid viability.
Collapse
Affiliation(s)
- Hong Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yuping Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jinsen Shi
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Ting Yan
- Department of General Surgery, Second Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jun Wang
- Department of General Surgery, Second Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yuning Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Qin Lu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Hailan Feng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jian Du
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Zhiyun Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| |
Collapse
|
9
|
Martinez-Ruiz L, López-Rodríguez A, Florido J, Rodríguez-Santana C, Rodríguez Ferrer JM, Acuña-Castroviejo D, Escames G. Patient-derived tumor models in cancer research: Evaluation of the oncostatic effects of melatonin. Biomed Pharmacother 2023; 167:115581. [PMID: 37748411 DOI: 10.1016/j.biopha.2023.115581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
The development of new anticancer therapies tends to be very slow. Although their impact on potential candidates is confirmed in preclinical studies, ∼95 % of these new therapies are not approved when tested in clinical trials. One of the main reasons for this is the lack of accurate preclinical models. In this context, there are different patient-derived models, which have emerged as a powerful oncological tool: patient-derived xenografts (PDXs), patient-derived organoids (PDOs), and patient-derived cells (PDCs). Although all these models are widely applied, PDXs, which are created by engraftment of patient tumor tissues into mice, is considered more reliable. In fundamental research, the PDX model is used to evaluate drug-sensitive markers and, in clinical practice, to select a personalized therapeutic strategy. Melatonin is of particular importance in the development of innovative cancer treatments due to its oncostatic impact and lack of adverse effects. However, the literature regarding the oncostatic effect of melatonin in patient-derived tumor models is scant. This review aims to describe the important role of patient-derived models in the development of anticancer treatments, focusing, in particular, on PDX models, as well as their use in cancer research. This review also summarizes the existing literature on the anti-tumoral effect of melatonin in patient-derived models in order to propose future anti-neoplastic clinical applications.
Collapse
Affiliation(s)
- Laura Martinez-Ruiz
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Alba López-Rodríguez
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Javier Florido
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Cesar Rodríguez-Santana
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - José M Rodríguez Ferrer
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Germaine Escames
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain.
| |
Collapse
|
10
|
Weng G, Tao J, Liu Y, Qiu J, Su D, Wang R, Luo W, Zhang T. Organoid: Bridging the gap between basic research and clinical practice. Cancer Lett 2023; 572:216353. [PMID: 37599000 DOI: 10.1016/j.canlet.2023.216353] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Nowadays, the diagnosis and treatment system of malignant tumors has increasingly tended to be more precise and personalized while the existing tumor models are still unable to fully meet the needs of clinical practice. Notably, the emerging organoid platform has been proven to have huge potential in the field of basic-translational medicine, which is expected to promote a paradigm shift in personalized medicine. Here, given the unique advantages of organoid platform, we mainly explore the prominent role of organoid models in basic research and clinical practice from perspectives of tumor biology, tumorigenic microbes-host interaction, clinical decision-making, and regenerative strategy. In addition, we also put forward some practical suggestions on how to construct a new generation of organoid platform, which is destined to vigorously promote the reform of basic-translational medicine.
Collapse
Affiliation(s)
- Guihu Weng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Dan Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Ruobing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
11
|
Koga T, Soh J, Hamada A, Miyano Y, Fujino T, Obata K, Ohara S, Nishino M, Chiba M, Shimoji M, Takemoto T, Suda K, Sakai K, Sato H, Mitsudomi T. Clinical Relevance of Patient-Derived Organoid of Surgically Resected Lung Cancer as an In Vitro Model for Biomarker and Drug Testing. JTO Clin Res Rep 2023; 4:100554. [PMID: 37681218 PMCID: PMC10480534 DOI: 10.1016/j.jtocrr.2023.100554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/01/2023] [Accepted: 07/21/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Lung tumor organoids (LTOs) have attracted attention as in vitro preclinical models; however, their clinical and experimental applications have not been fully established. Methods We attempted to establish LTOs from resected specimens of patients with lung cancer who underwent lung resection. Clinicopathologic characteristics related to the establishment of LTOs were evaluated. Histologic assessment and genetic analysis were conducted for both LTOs and their parental tumors. Organoid-derived xenografts were generated in immunocompetent mice. Drug sensitivity was assessed using cell proliferation assays. Results We established 53 LTOs from 79 lung cancer samples, including 10 long-term culture models. The establishment rate was significantly lower in squamous cell carcinomas than in other histologic types (48% versus 75%, p = 0.034). Histologic similarities were confirmed among LTOs, the parental tumors, and organoid-derived xenografts. Seven mutations, including two EGFR L858R and one EGFR exon 20 H773delinsYNPY mutations, were detected in both LTO and parental tumors; the other four mutations were detected in either LTO or parental tumors. The extensive culture ability of LTO (passaged >10 times) correlated with poor patient prognosis. LTO9 cells harboring EGFR H773delinsYNPY were sensitive to osimertinib. The parental patient, who had new metastatic lesions, was treated with osimertinib and exhibited a remarkable response. Conclusions The establishment and growth rates of LTOs were associated with the histologic subtype and tumor size. LTOs derived from resected specimens have become preclinical models that can be used to predict drug responses and accelerate the development of treatment strategies for patients with rare mutations.
Collapse
Affiliation(s)
- Takamasa Koga
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Junichi Soh
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Akira Hamada
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yuki Miyano
- Genome Informatics Unit, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Toshio Fujino
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Keiko Obata
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Shuta Ohara
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaya Nishino
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masato Chiba
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaki Shimoji
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Toshiki Takemoto
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenichi Suda
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hidenori Sato
- Genome Informatics Unit, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Kindai Hospital Global Research Alliance Center, Kindai University Hospital, Osaka-Sayama, Japan
| |
Collapse
|
12
|
Yang C, Xiao W, Wang R, Hu Y, Yi K, Sun X, Wang G, Xu X. Tumor organoid model of colorectal cancer (Review). Oncol Lett 2023; 26:328. [PMID: 37415635 PMCID: PMC10320425 DOI: 10.3892/ol.2023.13914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
The establishment of self-organizing 'mini-gut' organoid models has brought about a significant breakthrough in biomedical research. Patient-derived tumor organoids have emerged as valuable tools for preclinical studies, offering the retention of genetic and phenotypic characteristics of the original tumor. These organoids have applications in various research areas, including in vitro modelling, drug discovery and personalized medicine. The present review provided an overview of intestinal organoids, focusing on their unique characteristics and current understanding. The progress made in colorectal cancer (CRC) organoid models was then delved into, discussing their role in drug development and personalized medicine. For instance, it has been indicated that patient-derived tumor organoids are able to predict response to irinotecan-based neoadjuvant chemoradiotherapy. Furthermore, the limitations and challenges associated with current CRC organoid models were addressed, along with proposed strategies for enhancing their utility in future basic and translational research.
Collapse
Affiliation(s)
- Chi Yang
- Department of Gastroenterology, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| | - Wangwen Xiao
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| | - Rui Wang
- School of Pharmacy, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Yan Hu
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| | - Ke Yi
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| | - Xuan Sun
- Department of Gastroenterology, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| | - Guanghui Wang
- School of Pharmacy, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xiaohui Xu
- Department of Gastroenterology, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Soochow Medical College of Soochow University, Suzhou, Jiangsu 215400, P.R. China
| |
Collapse
|
13
|
Zeng X, Ma Q, Li XK, You LT, Li J, Fu X, You FM, Ren YF. Patient-derived organoids of lung cancer based on organoids-on-a-chip: enhancing clinical and translational applications. Front Bioeng Biotechnol 2023; 11:1205157. [PMID: 37304140 PMCID: PMC10250649 DOI: 10.3389/fbioe.2023.1205157] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Lung cancer is one of the most common malignant tumors worldwide, with high morbidity and mortality due to significant individual characteristics and genetic heterogeneity. Personalized treatment is necessary to improve the overall survival rate of the patients. In recent years, the development of patient-derived organoids (PDOs) enables lung cancer diseases to be simulated in the real world, and closely reflects the pathophysiological characteristics of natural tumor occurrence and metastasis, highlighting their great potential in biomedical applications, translational medicine, and personalized treatment. However, the inherent defects of traditional organoids, such as poor stability, the tumor microenvironment with simple components and low throughput, limit their further clinical transformation and applications. In this review, we summarized the developments and applications of lung cancer PDOs and discussed the limitations of traditional PDOs in clinical transformation. Herein, we looked into the future and proposed that organoids-on-a-chip based on microfluidic technology are advantageous for personalized drug screening. In addition, combined with recent advances in lung cancer research, we explored the translational value and future development direction of organoids-on-a-chip in the precision treatment of lung cancer.
Collapse
Affiliation(s)
- Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xue-Ke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Li-Ting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jia Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi-Feng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Miyazaki K, Xu C, Shimada M, Goel A. Curcumin and Andrographis Exhibit Anti-Tumor Effects in Colorectal Cancer via Activation of Ferroptosis and Dual Suppression of Glutathione Peroxidase-4 and Ferroptosis Suppressor Protein-1. Pharmaceuticals (Basel) 2023; 16:383. [PMID: 36986483 PMCID: PMC10055708 DOI: 10.3390/ph16030383] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Colorectal cancer (CRC) is the leading cause of cancer-related deaths worldwide. The limitations of current chemotherapeutic drugs in CRC include their toxicity, side effects, and exorbitant costs. To assess these unmet needs in CRC treatment, several naturally occurring compounds, including curcumin and andrographis, have gained increasing attention due to their multi-targeted functionality and safety vs. conventional drugs. In the current study, we revealed that a combination of curcumin and andrographis exhibited superior anti-tumor effects by inhibiting cell proliferation, invasion, colony formation, and inducing apoptosis. Genome-wide transcriptomic expression profiling analysis revealed that curcumin and andrographis activated the ferroptosis pathway. Moreover, we confirmed the gene and protein expression of glutathione peroxidase 4 (GPX-4) and ferroptosis suppressor protein 1 (FSP-1), the two major negative regulators of ferroptosis, were downregulated by this combined treatment. With this regimen, we also observed that intracellular accumulation of reactive oxygen species and lipid peroxides were induced in CRC cells. These cell line findings were validated in patient-derived organoids. In conclusion, our study revealed that combined treatment with curcumin and andrographis exhibited anti-tumorigenic effects in CRC cells through activation of ferroptosis and by dual suppression of GPX-4 and FSP-1, which have significant potential implications for the adjunctive treatment of CRC patients.
Collapse
Affiliation(s)
- Katsuki Miyazaki
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91016, USA
- Department of Surgery, Tokushima University, Tokushima 770-0042, Japan
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91016, USA
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116004, China
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima 770-0042, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91016, USA
| |
Collapse
|
15
|
Acharayothin O, Thiengtrong B, Juengwiwattanakitti P, Anekwiang P, Riansuwan W, Chinswangwatanakul V, Tanjak P. Impact of Washing Processes on RNA Quantity and Quality in Patient-Derived Colorectal Cancer Tissues. Biopreserv Biobank 2023; 21:31-37. [PMID: 35230139 DOI: 10.1089/bio.2021.0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Colorectal cancer (CRC) is a common and lethal cancer worldwide. Extraction of high-quality RNA from CRC samples plays a key role in scientific research and translational medicine. Specimen collection and washing methods that do not compromise RNA quality or quantity are needed to ensure high quality specimens for gene expression analysis and other RNA-based downstream applications. We investigated the effect of tissue specimen collection and different preparation processes on the quality and quantity of RNA extracted from surgical CRC tissues. Materials and Methods: After surgical resection, tissues were harvested and prepared with various washing processes in a room adjacent to the operating room. One hundred fourteen tissues from 36 CRC patients were separately washed in either cold phosphate-buffered saline reagent (n = 34) or Dulbecco's modified Eagle's medium (DMEM; n = 34) for 2-3 minutes until the stool was removed, and unwashed specimens served as controls (n = 34). Six tissue specimens were washed and immersed in DMEM for up to 1 hour at 4°C. Before RNA extraction, all specimens were kept in the stabilizing reagent for 3 months at -80°C. RNA was extracted, and the concentration per milligram of tissue was measured. RNA quality was assessed using the RNA integrity number (RIN) value. Results: Different washing processes did not result in significant differences in RNA quantity or RIN values. In the six tissues that were washed and immersed in DMEM for 1 hour, RIN values significantly decreased. The quality of the extracted RNA from most specimens was excellent with the average RIN greater than 7. Conclusions: RNA is stable in specimens washed in different processes for short periods, but RIN values may decrease with prolonged wash times.
Collapse
Affiliation(s)
- Onchira Acharayothin
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Benjarat Thiengtrong
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Panudeth Juengwiwattanakitti
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Panatna Anekwiang
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Woramin Riansuwan
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vitoon Chinswangwatanakul
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pariyada Tanjak
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
16
|
The In Vitro Adaptation of Patient-Derived Organoids Suggests Alternative Strategies against CMS1 Colorectal Cancer: When the Microenvironment Does Make the Difference. Cancers (Basel) 2022; 14:cancers14246086. [PMID: 36551571 PMCID: PMC9775687 DOI: 10.3390/cancers14246086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a relatively slow-growing tumor that can be treated successfully when identified in the early stages [...].
Collapse
|
17
|
Löwenmark T, Löfgren-Burström A, Zingmark C, Ljuslinder I, Dahlberg M, Edin S, Palmqvist R. Tumour Colonisation of Parvimonas micra Is Associated with Decreased Survival in Colorectal Cancer Patients. Cancers (Basel) 2022; 14:5937. [PMID: 36497419 PMCID: PMC9736682 DOI: 10.3390/cancers14235937] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Increasing evidence suggests that the gut microbiota may impact colorectal cancer (CRC) development and progression. In this study, the tumour colonisation of two CRC-associated bacteria, Parvimonas micra and Fusobacterium nucleatum, was studied in relation to patient survival in a cohort of 257 CRC patients. Colonisation of P. micra and F. nucleatum was analysed in fresh frozen tumour tissue (n = 112) and in faeces (n = 250) by qPCR. When analysing tumour tissues, both P. micra and F. nucleatum were found to be associated with decreased five-year cancer-specific survival, an association that remained significant in multivariable analysis for P. micra. Furthermore, we found significant associations of high levels of P. micra and F. nucleatum with tumour molecular characteristics, i.e., tumours mutated in BRAFV600E, and tumours of the MSI subtype. The analysis of faecal samples showed weaker associations with prognosis and tumour molecular characteristics. In conclusion, our findings support a novel association of tumour colonisation of P. micra with decreased patient survival. A better understanding of the role of the gut microbiota in CRC might contribute to the advancement of prognostic tools and new targets for therapy.
Collapse
Affiliation(s)
- Thyra Löwenmark
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| | - Anna Löfgren-Burström
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, SE-90185 Umeå, Sweden
| | - Michael Dahlberg
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, SE-90185 Umeå, Sweden
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| |
Collapse
|
18
|
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: 7] [Impact Index Per Article: 2.3] [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
|
19
|
Cioce M, Sacconi A, Donzelli S, Bonomo C, Perracchio L, Carosi M, Telera S, Fazio VM, Botti C, Strano S, Blandino G. Breast cancer metastasis: is it a matter of OMICS and proper ex-vivo models? Comput Struct Biotechnol J 2022; 20:4003-4008. [PMID: 35983233 PMCID: PMC9355905 DOI: 10.1016/j.csbj.2022.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Abstract
Genomics has greatly increased the understanding of the study of breast cancer (BC) and has shaped the concept of intra-tumor heterogeneity, currently recognized as a propelling force for cancer progression. In this context, knowledge and understanding of metastatic breast cancer (mBC) has somehow lagged behind that of primary breast cancer. This may be explained by the relative scarcity of matched mBC samples, however it is possible that the mutation spectrum obtained from primary BC does not capture the full complexity of the metastatic disease. Here, we provide a few examples supporting this possibility, from public databases. We evoke the need to perform an integrated multi-OMICS characterization of mBC, to obtain a broad understanding of this complex disease, whose evolution cannot be explained solely by genomics. Pertinent to this, we suggest that rather an infrequent use of Patient-Derived –Tumor-Organoids (PDTOs) may be influenced by assuming that the metastatic conditions of PDTOs growth (mPDTOs) should be similar to those of the tissue of origin. We challenge this view by suggesting that the use of “target-organ inspired” growth conditions for mPDTOs, may better fit the emerging knowledge of metastatic disease. Thus, the integrated use of multi-OMICS and of clinically relevant mPDTOs may allow a further understanding of such disease and foster therapeutically relevant advances. We believe that our points may be valid for other solid cancers.
Collapse
Affiliation(s)
- Mario Cioce
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Andrea Sacconi
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sara Donzelli
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Claudia Bonomo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Letizia Perracchio
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariantonia Carosi
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Telera
- Neurosurgery Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Vito Michele Fazio
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Claudio Botti
- Breast Surgery Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sabrina Strano
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Corresponding author at: Translational Oncology Research UnitItalian National Cancer Institute Regina Elena Rome, Italy.
| |
Collapse
|
20
|
Patient Derived Ex-Vivo Cancer Models in Drug Development, Personalized Medicine, and Radiotherapy. Cancers (Basel) 2022; 14:cancers14123006. [PMID: 35740672 PMCID: PMC9220792 DOI: 10.3390/cancers14123006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary This review article highlights gaps in the current system of drug development and personalized medicine for cancer therapy. The ex vivo model system using tissue biopsy from patients will advance the development of the predictive disease specific biomarker, drug screening and assessment of treatment response on a personalized basis. Although this ex vivo system demonstrated promises, there are challenges and limitations which need to be mitigated for further advancement and better applications. Abstract The field of cancer research is famous for its incremental steps in improving therapy. The consistent but slow rate of improvement is greatly due to its meticulous use of consistent cancer biology models. However, as we enter an era of increasingly personalized cancer care, including chemo and radiotherapy, our cancer models must be equally able to be applied to all individuals. Patient-derived organoid (PDO) and organ-in-chip (OIC) models based on the micro-physiological bioengineered platform have already been considered key components for preclinical and translational studies. Accounting for patient variability is one of the greatest challenges in the crossover from preclinical development to clinical trials and patient derived organoids may offer a steppingstone between the two. In this review, we highlight how incorporating PDO’s and OIC’s into the development of cancer therapy promises to increase the efficiency of our therapeutics.
Collapse
|
21
|
Lee HH, Chen CH, Huang YH, Chiang CH, Huang MY. Biomarkers of Favorable vs. Unfavorable Responses in Locally Advanced Rectal Cancer Patients Receiving Neoadjuvant Concurrent Chemoradiotherapy. Cells 2022; 11:cells11101611. [PMID: 35626648 PMCID: PMC9139800 DOI: 10.3390/cells11101611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer is the second leading cause of cancer death globally. The gold standard for locally advanced rectal cancer (LARC) nowadays is preoperative concurrent chemoradiation (CCRT). Approximately three quarters of LARC patients do not achieve pathological complete response and hence suffer from relapse, metastases and inevitable death. The exploration of trustworthy and timely biomarkers for CCRT response is urgently called for. This review focused upon a broad spectrum of biomarkers, including circulating tumor cells, DNA, RNA, oncogenes, tumor suppressor genes, epigenetics, impaired DNA mismatch repair, patient-derived xenografts, in vitro tumor organoids, immunity and microbiomes. Utilizing proper biomarkers can assist in categorizing appropriate patients by the most efficient treatment modality with the best outcome and accompanied by minimal side effects. The purpose of this review is to inspect and analyze accessible data in order to fully realize the promise of precision oncology for rectal cancer patients.
Collapse
Affiliation(s)
- Hsin-Hua Lee
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Hung Chen
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 801, Taiwan
| | - Yu-Hsiang Huang
- Post-Graduate Year Training, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Cheng-Han Chiang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
| | - Ming-Yii Huang
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-3121101 (ext. 7158)
| |
Collapse
|
22
|
Baião A, Dias S, Soares AF, Pereira CL, Oliveira C, Sarmento B. Advances in the use of 3D colorectal cancer models for novel drug discovery. Expert Opin Drug Discov 2022; 17:569-580. [PMID: 35343351 DOI: 10.1080/17460441.2022.2056162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is one of the most common and deadly tumors worldwide. CRC in vitro and in vivo models that recapitulate key features of human disease are essential to the development of novel and effective therapeutics. However, two-dimensional (2D) in vitro culture systems are considered too simple and do not represent the complex nature of the human tumor. However, three-dimensional (3D) models have emerged in recent years as more advanced and complex cell culture systems, able to closely resemble key features of human cancer tissues. AREAS COVERED The authors' review the currently established in vitro cell culture models and describe the advances in the development of 3D scaffold-free models to study CRC. The authors also discuss intestinal spheroids and organoids. As well as in vitro models for drug screening and metastatic CRC (mCRC). EXPERT OPINION The ideal CRC in vitro model is not yet established. Spheroid-based 3D models represent one of the most used approaches to recapitulate the tumor environment, overcoming some limitations of 2D models. Mouse and patient-derived organoids are more advanced models that can mimic more closely the characteristics and properties of CRC, with the possibility of including cells derived from patients with metastatic CRC.
Collapse
Affiliation(s)
- Ana Baião
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia Dias
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Ana Francisca Soares
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Catarina Leite Pereira
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Carla Oliveira
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,IPATIMUP, Institute of Molecular Pathology and Immunology of University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,Department of Pathology, Faculty of Medicine of University of Porto, 4200-319 Porto, Portugal
| | - Bruno Sarmento
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
23
|
Furbo S, Urbano PCM, Raskov HH, Troelsen JT, Kanstrup Fiehn AM, Gögenur I. Use of Patient-Derived Organoids as a Treatment Selection Model for Colorectal Cancer: A Narrative Review. Cancers (Basel) 2022; 14:cancers14041069. [PMID: 35205817 PMCID: PMC8870458 DOI: 10.3390/cancers14041069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is the third most common type of cancer globally. Despite successful treatment, it has a 40% chance of recurrence within five years after surgery. While neoadjuvant chemotherapy is offered for stage IV cancers, it comes with a risk of resistance and disease progression. CRC tumors vary biologically, recur frequently, and pose a significant risk for cancer-related mortality, making it increasingly relevant to develop methods to study personalized treatment. A tumor organoid is a miniature, multicellular, and 3D replica of a tumor in vitro that retains its characteristics. Here, we discuss the current methods of culturing organoids and the correlation of drug response in organoids with clinical responses in patients. This helps us to determine whether organoids can be used for treatment selection in a clinical setting. Based on the studies included, there was a strong correlation between treatment responses of organoids and clinical treatment responses. Abstract Surgical resection is the mainstay in intended curative treatment of colorectal cancer (CRC) and may be accompanied by adjuvant chemotherapy. However, 40% of the patients experience recurrence within five years of treatment, highlighting the importance of improved, personalized treatment options. Monolayer cell cultures and murine models, which are generally used to study the biology of CRC, are associated with certain drawbacks; hence, the use of organoids has been emerging. Organoids obtained from tumors display similar genotypic and phenotypic characteristics, making them ideal for investigating individualized treatment strategies and for integration as a core platform to be used in prediction models. Here, we review studies correlating the clinical response in patients with CRC with the therapeutic response in patient-derived organoids (PDO), as well as the limitations and potentials of this model. The studies outlined in this review reported strong associations between treatment responses in the PDO model and clinical treatment responses. However, as PDOs lack the tumor microenvironment, they do not genuinely account for certain crucial characteristics that influence therapeutic response. To this end, we reviewed studies investigating PDOs co-cultured with tumor-infiltrating lymphocytes. This model is a promising method allowing evaluation of patient-specific tumors and selection of personalized therapies. Standardized methodologies must be implemented to reach a “gold standard” for validating the use of this model in larger cohorts of patients. The introduction of this approach to a clinical scenario directing neoadjuvant treatment and in other curative and palliative treatment strategies holds incredible potential for improving personalized treatment and its outcomes.
Collapse
Affiliation(s)
- Sara Furbo
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (S.F.); (P.C.M.U.); (H.H.R.); (A.-M.K.F.)
| | - Paulo César Martins Urbano
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (S.F.); (P.C.M.U.); (H.H.R.); (A.-M.K.F.)
| | - Hans Henrik Raskov
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (S.F.); (P.C.M.U.); (H.H.R.); (A.-M.K.F.)
| | - Jesper Thorvald Troelsen
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark;
- Enhanced Perioperative Oncology (EPeOnc) Consortium, Zealand University Hospital, 4600 Køge, Denmark
| | - Anne-Marie Kanstrup Fiehn
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (S.F.); (P.C.M.U.); (H.H.R.); (A.-M.K.F.)
- Department of Pathology, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 København, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (S.F.); (P.C.M.U.); (H.H.R.); (A.-M.K.F.)
- Enhanced Perioperative Oncology (EPeOnc) Consortium, Zealand University Hospital, 4600 Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 København, Denmark
- Correspondence: ; Tel.: +45-2633-6426
| |
Collapse
|
24
|
Marinucci M, Ercan C, Taha-Mehlitz S, Fourie L, Panebianco F, Bianco G, Gallon J, Staubli S, Soysal SD, Zettl A, Rauthe S, Vosbeck J, Droeser RA, Bolli M, Peterli R, von Flüe M, Ng CKY, Kollmar O, Coto-Llerena M, Piscuoglio S. Standardizing Patient-Derived Organoid Generation Workflow to Avoid Microbial Contamination From Colorectal Cancer Tissues. Front Oncol 2022; 11:781833. [PMID: 35083141 PMCID: PMC8784867 DOI: 10.3389/fonc.2021.781833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
The use of patient-derived organoids (PDO) as a valuable alternative to in vivo models significantly increased over the last years in cancer research. The ability of PDOs to genetically resemble tumor heterogeneity makes them a powerful tool for personalized drug screening. Despite the extensive optimization of protocols for the generation of PDOs from colorectal tissue, there is still a lack of standardization of tissue handling prior to processing, leading to microbial contamination of the organoid culture. Here, using a cohort of 16 patients diagnosed with colorectal carcinoma (CRC), we aimed to test the efficacy of phosphate-buffered saline (PBS), penicillin/streptomycin (P/S), and Primocin, alone or in combination, in preventing organoid cultures contamination when used in washing steps prior to tissue processing. Each CRC tissue was divided into 5 tissue pieces, and treated with each different washing solution, or none. After the washing steps, all samples were processed for organoid generation following the same standard protocol. We detected contamination in 62.5% of the non-washed samples, while the use of PBS or P/S-containing PBS reduced the contamination rate to 50% and 25%, respectively. Notably, none of the organoid cultures washed with PBS/Primocin-containing solution were contaminated. Interestingly, addition of P/S to the washing solution reduced the percentage of living cells compared to Primocin. Taken together, our results demonstrate that, prior to tissue processing, adding Primocin to the tissue washing solution is able to eliminate the risk of microbial contamination in PDO cultures, and that the use of P/S negatively impacts organoids growth. We believe that our easy-to-apply protocol might help increase the success rate of organoid generation from CRC patients.
Collapse
Affiliation(s)
- Mattia Marinucci
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Caner Ercan
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Stephanie Taha-Mehlitz
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Lana Fourie
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Federica Panebianco
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gaia Bianco
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - John Gallon
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sebastian Staubli
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Savas D Soysal
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Andreas Zettl
- Institute of Pathology, Viollier AG, Allschwil, Switzerland
| | - Stephan Rauthe
- Institute of Pathology, Viollier AG, Allschwil, Switzerland
| | - Jürg Vosbeck
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Raoul A Droeser
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Martin Bolli
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Ralph Peterli
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Markus von Flüe
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Charlotte K Y Ng
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Otto Kollmar
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Basel, Switzerland
| | - Mairene Coto-Llerena
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
25
|
Wang J, Feng X, Li Z, Chen Y, Huang W. Patient-derived organoids as a model for tumor research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:259-326. [PMID: 35595351 DOI: 10.1016/bs.pmbts.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer represents a leading cause of death, despite the rapid progress of cancer research, leading to urgent need for accurate preclinical model to further study of tumor mechanism and accelerate translational applications. Cancer cell lines cannot fully recapitulate tumors of different patients due to the lack of tumor complexity and specification, while the high technical difficulty, long time, and substantial cost of patient-derived xenograft model makes it unable to be used extensively for all types of tumors and large-scale drug screening. Patient-derived organoids can be established rapidly with a high success rate from many tumors, and precisely replicate the key histopathological, genetic, and phenotypic features, as well as therapeutic response of patient tumor. Therefore, they are extensively used in cancer basic research, biobanking, disease modeling and precision medicine. The combinations of cancer organoids with other advanced technologies, such as 3D bio-printing, organ-on-a-chip, and CRISPR-Cas9, contributes to the more complete replication of complex tumor microenvironment and tumorigenesis. In this review, we discuss the various methods of the establishment and the application of patient-derived organoids in diverse tumors as well as the limitations and future prospects of these models. Further advances of tumor organoids are expected to bridge the huge gap between bench and bedside and provide the unprecedented opportunities to advance cancer research.
Collapse
Affiliation(s)
- Jia Wang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Xiaoying Feng
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China
| | - Yongsong Chen
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Weiren Huang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China; Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
26
|
Ebrahimi N, Nasr Esfahani A, Samizade S, Mansouri A, Ghanaatian M, Adelian S, Shadman Manesh V, Hamblin MR. The potential application of organoids in breast cancer research and treatment. Hum Genet 2021; 141:193-208. [PMID: 34713317 DOI: 10.1007/s00439-021-02390-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/16/2021] [Indexed: 12/23/2022]
Abstract
Tumor heterogeneity is a major challenge for breast cancer researchers who have struggled to find effective treatments despite recent advances in oncology. Although the use of 2D cell culture methods in breast cancer research has been effective, it cannot model the heterogeneity of breast cancer as found within the body. The development of 3D culture of tumor cells and breast cancer organoids has provided a new approach in breast cancer research, allowing the identification of biomarkers, study of the interaction of tumor cells with the microenvironment, and for drug screening and discovery. In addition, the possibility of gene editing in organoids, especially using the CRISPR/Cas9 system, is convenient, and has allowed a more detailed study of tumor behavior in models closer to the physiological condition. The present review covers the application of organoids in breast cancer research. The recent use of gene-editing systems to provide insights into therapeutic approaches for breast cancer, is highlighted. The study of organoids and the possibility of gene manipulation may be a step towards the personalized treatment of breast cancer, which has so far remained unattainable due to the high heterogeneity of breast cancer.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Division of Genetics, Department of Cell, Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Alireza Nasr Esfahani
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Setare Samizade
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Atena Mansouri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Ghanaatian
- Department of Microbiology, Islamic Azad University of Jahrom, Jahrom, Fars, Iran
| | - Samaneh Adelian
- Department of Genetics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vida Shadman Manesh
- Medical Engineering Tissue Engineering, Department of Medical Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Michael R Hamblin
- Faculty of Health Science, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028, South Africa.
| |
Collapse
|
27
|
Biomarkers and cell-based models to predict the outcome of neoadjuvant therapy for rectal cancer patients. Biomark Res 2021; 9:60. [PMID: 34321074 PMCID: PMC8317379 DOI: 10.1186/s40364-021-00313-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Rectal cancer constitutes approximately one-third of all colorectal cancers and contributes to considerable mortality globally. In contrast to colon cancer, the standard treatment for localized rectal cancer often involves neoadjuvant chemoradiotherapy. Tumour response rates to treatment show substantial inter-patient heterogeneity, indicating a need for treatment stratification. Consequently researchers have attempted to establish new means for predicting tumour response in order to assist in treatment decisions. In this review we have summarized published findings regarding potential biomarkers to predict neoadjuvant treatment response for rectal cancer tumours. In addition, we describe cell-based models that can be utilized both for treatment prediction and for studying the complex mechanisms involved.
Collapse
|
28
|
Durinikova E, Buzo K, Arena S. Preclinical models as patients' avatars for precision medicine in colorectal cancer: past and future challenges. J Exp Clin Cancer Res 2021; 40:185. [PMID: 34090508 PMCID: PMC8178911 DOI: 10.1186/s13046-021-01981-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a complex and heterogeneous disease, characterized by dismal prognosis and low survival rate in the advanced (metastatic) stage. During the last decade, the establishment of novel preclinical models, leading to the generation of translational discovery and validation platforms, has opened up a new scenario for the clinical practice of CRC patients. To bridge the results developed at the bench with the medical decision process, the ideal model should be easily scalable, reliable to predict treatment responses, and flexibly adapted for various applications in the research. As such, the improved benefit of novel therapies being tested initially on valuable and reproducible preclinical models would lie in personalized treatment recommendations based on the biology and genomics of the patient's tumor with the overall aim to avoid overtreatment and unnecessary toxicity. In this review, we summarize different in vitro and in vivo models, which proved efficacy in detection of novel CRC culprits and shed light into the biology and therapy of this complex disease. Even though cell lines and patient-derived xenografts remain the mainstay of colorectal cancer research, the field has been confidently shifting to the use of organoids as the most relevant preclinical model. Prioritization of organoids is supported by increasing body of evidence that these represent excellent tools worth further therapeutic explorations. In addition, novel preclinical models such as zebrafish avatars are emerging as useful tools for pharmacological interrogation. Finally, all available models represent complementary tools that can be utilized for precision medicine applications.
Collapse
Affiliation(s)
- Erika Durinikova
- Candiolo Cancer Institute, FPO - IRCCS, Strada Provinciale 142, Km 3.95, 10060, Candiolo, TO, Italy
| | - Kristi Buzo
- Candiolo Cancer Institute, FPO - IRCCS, Strada Provinciale 142, Km 3.95, 10060, Candiolo, TO, Italy
| | - Sabrina Arena
- Candiolo Cancer Institute, FPO - IRCCS, Strada Provinciale 142, Km 3.95, 10060, Candiolo, TO, Italy.
- Department of Oncology, University of Torino, Strada Provinciale 142, Km 3.95, 10060, Candiolo, TO, Italy.
| |
Collapse
|
29
|
Organoids and Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13112657. [PMID: 34071313 PMCID: PMC8197877 DOI: 10.3390/cancers13112657] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids were first established as a three-dimensional cell culture system from mouse small intestine. Subsequent development has made organoids a key system to study many human physiological and pathological processes that affect a variety of tissues and organs. In particular, organoids are becoming very useful tools to dissect colorectal cancer (CRC) by allowing the circumvention of classical problems and limitations, such as the impossibility of long-term culture of normal intestinal epithelial cells and the lack of good animal models for CRC. In this review, we describe the features and current knowledge of intestinal organoids and how they are largely contributing to our better understanding of intestinal cell biology and CRC genetics. Moreover, recent data show that organoids are appropriate systems for antitumoral drug testing and for the personalized treatment of CRC patients.
Collapse
|
30
|
Pramil E, Dillard C, Escargueil AE. Colorectal Cancer and Immunity: From the Wet Lab to Individuals. Cancers (Basel) 2021; 13:cancers13071713. [PMID: 33916641 PMCID: PMC8038567 DOI: 10.3390/cancers13071713] [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: 03/01/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Tackling the current dilemma of colorectal cancer resistance to immunotherapy is puzzling and requires novel therapeutic strategies to emerge. However, characterizing the intricate interactions between cancer and immune cells remains difficult because of the complexity and heterogeneity of both compartments. Developing rationales is intellectually feasible but testing them can be experimentally challenging and requires the development of innovative procedures and protocols. In this review, we delineated useful in vitro and in vivo models used for research in the field of immunotherapy that are or could be applied to colorectal cancer management and lead to major breakthroughs in the coming years. Abstract Immunotherapy is a very promising field of research and application for treating cancers, in particular for those that are resistant to chemotherapeutics. Immunotherapy aims at enhancing immune cell activation to increase tumor cells recognition and killing. However, some specific cancer types, such as colorectal cancer (CRC), are less responsive than others to the current immunotherapies. Intrinsic resistance can be mediated by the development of an immuno-suppressive environment in CRC. The mutational status of cancer cells also plays a role in this process. CRC can indeed be distinguished in two main subtypes. Microsatellite instable (MSI) tumors show a hyper-mutable phenotype caused by the deficiency of the DNA mismatch repair machinery (MMR) while microsatellite stable (MSS) tumors show a comparatively more “stable” mutational phenotype. Several studies demonstrated that MSI CRC generally display good prognoses for patients and immunotherapy is considered as a therapeutic option for this type of tumors. On the contrary, MSS metastatic CRC usually presents a worse prognosis and is not responsive to immunotherapy. According to this, developing new and innovative models for studying CRC response towards immune targeted therapies has become essential in the last years. Herein, we review the in vitro and in vivo models used for research in the field of immunotherapy applied to colorectal cancer.
Collapse
Affiliation(s)
- Elodie Pramil
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Alliance Pour la Recherche en Cancérologie—APREC, Tenon Hospital, F-75012 Paris, France
| | - Clémentine Dillard
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Alliance Pour la Recherche en Cancérologie—APREC, Tenon Hospital, F-75012 Paris, France
| | - Alexandre E. Escargueil
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Correspondence: ; Tel.: +33-(0)1-49-28-46-44
| |
Collapse
|
31
|
Witte KE, Hertel O, Windmöller BA, Helweg LP, Höving AL, Knabbe C, Busche T, Greiner JFW, Kalinowski J, Noll T, Mertzlufft F, Beshay M, Pfitzenmaier J, Kaltschmidt B, Kaltschmidt C, Banz-Jansen C, Simon M. Nanopore Sequencing Reveals Global Transcriptome Signatures of Mitochondrial and Ribosomal Gene Expressions in Various Human Cancer Stem-like Cell Populations. Cancers (Basel) 2021; 13:cancers13051136. [PMID: 33800955 PMCID: PMC7962028 DOI: 10.3390/cancers13051136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer is the leading cause of death in the industrialized world. In particular, so-called cancer stem cells (CSCs) play a crucial role in disease progression, as they are known to contribute to tumor growth and metastasis. Thus, CSCs are heavily investigated in a broad range of cancers. Nevertheless, global transcriptomic profiling of CSC populations derived from different tumor types is rare. We established three CSC populations from tumors in the uterus, brain, lung, and prostate and assessed their global transcriptomes using nanopore full-length cDNA sequencing, a new technique to assess insights into global gene profile. We observed common expression in all CSCs for distinct genes encoding proteins for organelles, such as ribosomes, mitochondria, and proteasomes. Additionally, we detected high expressions of inflammation- and immunity-related genes. Conclusively, we observed high similarities between all CSCs independent of their tumor of origin, which may build the basis for identifying novel therapeutic strategies targeting CSCs. Abstract Cancer stem cells (CSCs) are crucial mediators of tumor growth, metastasis, therapy resistance, and recurrence in a broad variety of human cancers. Although their biology is increasingly investigated within the distinct types of cancer, direct comparisons of CSCs from different tumor types allowing comprehensive mechanistic insights are rarely assessed. In the present study, we isolated CSCs from endometrioid carcinomas, glioblastoma multiforme as well as adenocarcinomas of lung and prostate and assessed their global transcriptomes using full-length cDNA nanopore sequencing. Despite the expression of common CSC markers, principal component analysis showed a distinct separation of the CSC populations into three clusters independent of the specific type of tumor. However, GO-term and KEGG pathway enrichment analysis revealed upregulated genes related to ribosomal biosynthesis, the mitochondrion, oxidative phosphorylation, and glycolytic pathways, as well as the proteasome, suggesting a great extent of metabolic flexibility in CSCs. Interestingly, the GO term “NF-kB binding” was likewise found to be elevated in all investigated CSC populations. In summary, we here provide evidence for high global transcriptional similarities between CSCs from various tumors, which particularly share upregulated gene expression associated with mitochondrial and ribosomal activity. Our findings may build the basis for identifying novel therapeutic strategies targeting CSCs.
Collapse
Affiliation(s)
- Kaya E. Witte
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Correspondence: ; Tel.: +49-521-106-5629
| | - Oliver Hertel
- Department of Cell Culture Technology, Faculty of Technology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (O.H.); (T.N.)
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Beatrice A. Windmöller
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Laureen P. Helweg
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Anna L. Höving
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Cornelius Knabbe
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Tobias Busche
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Jörn Kalinowski
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Thomas Noll
- Department of Cell Culture Technology, Faculty of Technology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (O.H.); (T.N.)
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Fritz Mertzlufft
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Scientific Director of the Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Maraweg 21, 33699 Bielefeld, Germany
| | - Morris Beshay
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department for Thoracic Surgery and Pneumology, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Jesco Pfitzenmaier
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Urology and Center for Computer-Assisted and Robotic Urology, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, Universitätsstrasse 25, 33699 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Constanze Banz-Jansen
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Matthias Simon
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Neurosurgery and Epilepsy Surgery, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| |
Collapse
|
32
|
Duzagac F, Saorin G, Memeo L, Canzonieri V, Rizzolio F. Microfluidic Organoids-on-a-Chip: Quantum Leap in Cancer Research. Cancers (Basel) 2021; 13:737. [PMID: 33578886 PMCID: PMC7916612 DOI: 10.3390/cancers13040737] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Organ-like cell clusters, so-called organoids, which exhibit self-organized and similar organ functionality as the tissue of origin, have provided a whole new level of bioinspiration for ex vivo systems. Microfluidic organoid or organs-on-a-chip platforms are a new group of micro-engineered promising models that recapitulate 3D tissue structure and physiology and combines several advantages of current in vivo and in vitro models. Microfluidics technology is used in numerous applications since it allows us to control and manipulate fluid flows with a high degree of accuracy. This system is an emerging tool for understanding disease development and progression, especially for personalized therapeutic strategies for cancer treatment, which provide well-grounded, cost-effective, powerful, fast, and reproducible results. In this review, we highlight how the organoid-on-a-chip models have improved the potential of efficiency and reproducibility of organoid cultures. More widely, we discuss current challenges and development on organoid culture systems together with microfluidic approaches and their limitations. Finally, we describe the recent progress and potential utilization in the organs-on-a-chip practice.
Collapse
Affiliation(s)
- Fahriye Duzagac
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy;
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
33
|
Reidy E, Leonard NA, Treacy O, Ryan AE. A 3D View of Colorectal Cancer Models in Predicting Therapeutic Responses and Resistance. Cancers (Basel) 2021; 13:E227. [PMID: 33435170 PMCID: PMC7827038 DOI: 10.3390/cancers13020227] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Although there have been many advances in recent years for the treatment of colorectal cancer (CRC), it still remains the third most common cause of cancer-related deaths worldwide. Many patients with late stage CRC display resistance to multiple different therapeutics. An important aspect in developing effective therapeutics for CRC patients is understanding the interactions that take place in the tumor microenvironment (TME), as it has been shown to contribute to drug resistance in vivo. Much research over the past 100 years has focused on 2D monolayer cultures or in vivo studies, however, the efficacy in translating these to the clinic is very low. More recent studies are turning towards developing an effective 3D model of CRC that is clinically relevant, that can recapitulate the TME in vitro and bridge the gap between 2D cultures and in vivo studies, with the aim of reducing the use of animal models in the future. This review summarises the advantages and limitations of different 3D CRC models. It emphasizes how different 3D models may be optimised to study cellular and extracellular interactions that take place in the TME of CRC in an effort to allow the development of more translatable effective treatment options for patients.
Collapse
Affiliation(s)
- Eileen Reidy
- Lambe Institute for Translational research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland; (E.R.); (N.A.L.); (O.T.)
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W5P7 Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, NUI Galway, H91 W2TY Galway, Ireland
| | - Niamh A. Leonard
- Lambe Institute for Translational research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland; (E.R.); (N.A.L.); (O.T.)
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W5P7 Galway, Ireland
| | - Oliver Treacy
- Lambe Institute for Translational research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland; (E.R.); (N.A.L.); (O.T.)
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W5P7 Galway, Ireland
| | - Aideen E. Ryan
- Lambe Institute for Translational research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland; (E.R.); (N.A.L.); (O.T.)
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W5P7 Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, NUI Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
34
|
Campaner E, Zannini A, Santorsola M, Bonazza D, Bottin C, Cancila V, Tripodo C, Bortul M, Zanconati F, Schoeftner S, Del Sal G. Breast Cancer Organoids Model Patient-Specific Response to Drug Treatment. Cancers (Basel) 2020; 12:E3869. [PMID: 33371412 PMCID: PMC7770601 DOI: 10.3390/cancers12123869] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor organoids are tridimensional cell culture systems that are generated in vitro from surgically resected patients' tumors. They can be propagated in culture maintaining several features of the tumor of origin, including cellular and genetic heterogeneity, thus representing a promising tool for precision cancer medicine. Here, we established patient-derived tumor organoids (PDOs) from different breast cancer subtypes (luminal A, luminal B, human epidermal growth factor receptor 2 (HER2)-enriched, and triple negative). The established model systems showed histological and genomic concordance with parental tumors. However, in PDOs, the ratio of diverse cell populations was frequently different from that originally observed in parental tumors. We showed that tumor organoids represent a valuable system to test the efficacy of standard therapeutic treatments and to identify drug resistant populations within tumors. We also report that inhibitors of mechanosignaling and of Yes-associated protein 1 (YAP) activation can restore chemosensitivity in drug resistant tumor organoids.
Collapse
Affiliation(s)
- Elena Campaner
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (E.C.); (A.Z.); (M.S.); (S.S.)
- National Laboratory CIB (LNCIB), Area Science Park—Padriciano, 34149 Trieste, Italy
| | - Alessandro Zannini
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (E.C.); (A.Z.); (M.S.); (S.S.)
- National Laboratory CIB (LNCIB), Area Science Park—Padriciano, 34149 Trieste, Italy
| | - Mariangela Santorsola
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (E.C.); (A.Z.); (M.S.); (S.S.)
- National Laboratory CIB (LNCIB), Area Science Park—Padriciano, 34149 Trieste, Italy
| | - Deborah Bonazza
- Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (D.B.); (C.B.); (M.B.); (F.Z.)
- UCO Anatomia e Istologia Patologica, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Hospital of Cattinara, 34149 Trieste, Italy
| | - Cristina Bottin
- Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (D.B.); (C.B.); (M.B.); (F.Z.)
- UCO Anatomia e Istologia Patologica, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Hospital of Cattinara, 34149 Trieste, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, School of Medicine, University of Palermo, 90133 Palermo, Italy; (V.C.); (C.T.)
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, School of Medicine, University of Palermo, 90133 Palermo, Italy; (V.C.); (C.T.)
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milano, Italy
| | - Marina Bortul
- Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (D.B.); (C.B.); (M.B.); (F.Z.)
- Breast Unit, Division of General Surgery, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Hospital of Cattinara, 34149 Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (D.B.); (C.B.); (M.B.); (F.Z.)
- UCO Anatomia e Istologia Patologica, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Hospital of Cattinara, 34149 Trieste, Italy
| | - Stefan Schoeftner
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (E.C.); (A.Z.); (M.S.); (S.S.)
- National Laboratory CIB (LNCIB), Area Science Park—Padriciano, 34149 Trieste, Italy
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (E.C.); (A.Z.); (M.S.); (S.S.)
- National Laboratory CIB (LNCIB), Area Science Park—Padriciano, 34149 Trieste, Italy
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milano, Italy
| |
Collapse
|
35
|
Current and Future Perspectives of the Use of Organoids in Radiobiology. Cells 2020; 9:cells9122649. [PMID: 33317153 PMCID: PMC7764598 DOI: 10.3390/cells9122649] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
Collapse
|
36
|
Onfroy-Roy L, Hamel D, Foncy J, Malaquin L, Ferrand A. Extracellular Matrix Mechanical Properties and Regulation of the Intestinal Stem Cells: When Mechanics Control Fate. Cells 2020; 9:cells9122629. [PMID: 33297478 PMCID: PMC7762382 DOI: 10.3390/cells9122629] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Intestinal stem cells (ISC) are crucial players in colon epithelium physiology. The accurate control of their auto-renewal, proliferation and differentiation capacities provides a constant flow of regeneration, maintaining the epithelial intestinal barrier integrity. Under stress conditions, colon epithelium homeostasis in disrupted, evolving towards pathologies such as inflammatory bowel diseases or colorectal cancer. A specific environment, namely the ISC niche constituted by the surrounding mesenchymal stem cells, the factors they secrete and the extracellular matrix (ECM), tightly controls ISC homeostasis. Colon ECM exerts physical constraint on the enclosed stem cells through peculiar topography, stiffness and deformability. However, little is known on the molecular and cellular events involved in ECM regulation of the ISC phenotype and fate. To address this question, combining accurately reproduced colon ECM mechanical parameters to primary ISC cultures such as organoids is an appropriated approach. Here, we review colon ECM physical properties at physiological and pathological states and their bioengineered in vitro reproduction applications to ISC studies.
Collapse
Affiliation(s)
- Lauriane Onfroy-Roy
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- Correspondence: (L.O.-R.); (A.F.); Tel.: +33-5-62-744-522 (A.F.)
| | - Dimitri Hamel
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Julie Foncy
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Laurent Malaquin
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Audrey Ferrand
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- Correspondence: (L.O.-R.); (A.F.); Tel.: +33-5-62-744-522 (A.F.)
| |
Collapse
|
37
|
Li X, Ling A, Kellgren TG, Lundholm M, Löfgren-Burström A, Zingmark C, Rutegård M, Ljuslinder I, Palmqvist R, Edin S. A Detailed Flow Cytometric Analysis of Immune Activity Profiles in Molecular Subtypes of Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12113440. [PMID: 33228141 PMCID: PMC7699331 DOI: 10.3390/cancers12113440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Colorectal cancer is one of the deadliest cancers worldwide, with around 40% of patients dying from distant metastasis. Tumour immune cell infiltration has powerful positive prognostic value in this disease, suggesting immunotherapy as a potential treatment modality. The aim of this explorative study was to assess in detail the local and systemic immune response in different molecular subgroups of colorectal cancer. An improved molecular understanding of the disease may lead to important advances in personalised medicine, identifying prognostic and predictive tools, in addition to new therapeutic targets. Abstract The local anti-tumour immune response has important prognostic value in colorectal cancer (CRC). In the era of immunotherapy, a better understanding of the immune response in molecular subgroups of CRC may lead to significant advances in personalised medicine. On this note, microsatellite instable (MSI) tumours have been characterised by increased immune infiltration, suggesting MSI as a marker for immune inhibitor checkpoint therapy. Here, we used flow cytometry to perform a comprehensive analysis of immune activity profiles in tumour tissues, adjacent non-malignant tissues and blood, from a cohort of 69 CRC patients. We found several signs of immune suppression in tumours compared to adjacent non-malignant tissues, including T cells more often expressing the immune checkpoint molecules programmed cell death protein (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). We further analysed immune cell infiltration in molecular subgroups of CRC. MSI tumours were indeed found to be associated with increased immune infiltration, including increased fractions of PD-1+ T cells. No correlation was, however, found between MSI and the fraction of CTLA-4+ T cells. Interestingly, within the group of patients with microsatellite stable (MSS) tumours, some also presented with increased immune infiltration, including comparably high portions of PD-1+ T cells, but also CTLA-4+ T cells. Furthermore, no correlation was found between PD-1+ and CTLA-4+ T cells, suggesting that different tumours may, to some extent, be regulated by different immune checkpoints. We further evaluated the distribution of immune activity profiles in the consensus molecular subtypes of CRC. In conclusion, our findings suggest that different immune checkpoint inhibitors may be beneficial for selected CRC patients irrespective of MSI status. Improved predictive tools are required to identify these patients.
Collapse
Affiliation(s)
- Xingru Li
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Agnes Ling
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Therese G. Kellgren
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Marie Lundholm
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Anna Löfgren-Burström
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Martin Rutegård
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, 90185 Umeå, Sweden;
- Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden;
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (A.L.); (T.G.K.); (M.L.); (A.L.-B.); (C.Z.); (R.P.)
- Correspondence: ; Tel.: +46-(0)907854431; Fax: +46-(0)90-121562
| |
Collapse
|
38
|
Panoutsopoulos AA. Organoids, Assembloids, and Novel Biotechnology: Steps Forward in Developmental and Disease-Related Neuroscience. Neuroscientist 2020; 27:463-472. [PMID: 32981451 DOI: 10.1177/1073858420960112] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In neuroscience research, the efforts to find the model through which we can mimic the in vivo microenvironment of a developing or defective brain have been everlasting. While model organisms are used for over a hundred years, many more methods have been introduced with immortalized or primary cell lines and later induced pluripotent stem cells and organoids to be some of these. As the use of organoids becomes more and more common by many laboratories in biology and neuroscience in particular, it is crucial to deeper understand the challenges and possible pitfalls of their application in research, many of which can be surpassed with the support of state-of-the art bioengineering solutions. In this review, after a brief chronicle of the path to the discovery of organoids, we focus on the latest approaches to study neuroscience related topics with organoids, such as the use of assembloids, CRISPR technology, patch-clamp and optogenetics techniques and discuss how modern 3-dimensional biomaterials, miniaturized bioreactors and microfluidic chips can help to overcome the disadvantages of their use.
Collapse
Affiliation(s)
- Alexios A Panoutsopoulos
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children-Northern California, Sacramento, CA, USA
| |
Collapse
|
39
|
Ye P, Xi Y, Huang Z, Xu P. Linking Obesity with Colorectal Cancer: Epidemiology and Mechanistic Insights. Cancers (Basel) 2020; 12:cancers12061408. [PMID: 32486076 PMCID: PMC7352519 DOI: 10.3390/cancers12061408] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
The incidence of obesity and colorectal cancer (CRC) has risen rapidly in recent decades. More than 650 million obese and 2 billion overweight individuals are currently living in the world. CRC is the third most common cancer. Obesity is regarded as one of the key environmental risk factors for the pathogenesis of CRC. In the present review, we mainly focus on the epidemiology of obesity and CRC in the world, the United States, and China. We also summarize the molecular mechanisms linking obesity to CRC in different aspects, including nutriology, adipokines and hormones, inflammation, gut microbiota, and bile acids. The unmet medical needs for obesity-related CRC are still remarkable. Understanding the molecular basis of these associations will help develop novel therapeutic targets and approaches for the treatment of obesity-related CRC.
Collapse
Affiliation(s)
- Pengfei Ye
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China;
| | - Yue Xi
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China;
| | - Zhiying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China;
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Correspondence: ; Tel.: +1-412-708-4694
| |
Collapse
|