1
|
De Ieso ML, Aldoghachi AF, Tilley WD, Dwyer AR. Are androgen receptor agonists a treatment option in bladder cancer? J Steroid Biochem Mol Biol 2025; 245:106623. [PMID: 39306143 DOI: 10.1016/j.jsbmb.2024.106623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 07/27/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Sex-related differences in bladder cancer incidence and progression infer a role for sex hormones and their cognate receptors in this disease. In part due to the oncogenic role of androgen receptor signaling in prostate cancer, the focus of most preclinical and clinical research to-date has been on the potential pro-tumorigenic action of androgens in urothelial cancers. However, clinical studies of androgen receptor antagonism have yielded minimal success. In this review, we explore the tumor suppressor role of androgen receptor in bladder cancer and discuss how it might be harnessed therapeutically.
Collapse
Affiliation(s)
- Michael L De Ieso
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Ahmed Faris Aldoghachi
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Amy R Dwyer
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
2
|
Taurin S, Alzahrani R, Aloraibi S, Ashi L, Alharmi R, Hassani N. Patient-derived tumor organoids: A preclinical platform for personalized cancer therapy. Transl Oncol 2025; 51:102226. [PMID: 39622151 DOI: 10.1016/j.tranon.2024.102226] [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: 09/13/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/11/2024] Open
Abstract
Patient-derived tumor organoids (PDTOs) represent a significant advancement in cancer research and personalized medicine. These organoids, derived from various cancer types, have shown the ability to retain the genetic and molecular characteristics of the original tumors, allowing for the detailed study of tumor biology and drug responses on an individual basis. The success rates of establishing PDTOs vary widely and are influenced by factors such as cancer type, tissue quality, and media composition. Furthermore, the dynamic nature of organoid cultures may also lead to unique molecular characteristics that deviate from the original tumors, affecting their interpretation in clinical settings without the implementation of rigorous validation and establishment of standardized protocols. Recent studies have supported the correlation between PDTOs and the corresponding patient response. Although these studies involved a small number of patients, they promoted the integration of PDTOs in observational and interventional clinical trials to advance translational cancer therapies.
Collapse
Affiliation(s)
- Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain.
| | - Reem Alzahrani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Sahar Aloraibi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Layal Ashi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Rawan Alharmi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Noora Hassani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
3
|
Li P, Huang M, Ma Y, Zhang Y, Shi C. Novel research model for in vitro immunotherapy: co-culturing tumor organoids with peripheral blood mononuclear cells. Cancer Cell Int 2024; 24:438. [PMID: 39741287 DOI: 10.1186/s12935-024-03628-3] [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: 04/01/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025] Open
Abstract
Tumor organoids have emerged as powerful tools for in vitro cancer research due to their ability to retain the structural and genetic characteristics of tumors. Nevertheless, the absence of a complete tumor microenvironment (TME) limits the broader application of organoid models in immunological studies. Given the critical role of immune cells in tumor initiation and progression, the co-culture model of organoids and peripheral blood mononuclear cells (PBMCs) may provide an effective platform for simulating the interactions between immune and tumor cells in vitro. This model stands as a robust instrument for dissecting the TME, elucidating the molecular interactions, and exploring the therapeutic applications of chimeric antigen receptor (CAR)-engineered lymphocytes, as well as other cancer treatment modalities. This review systematically evaluates the advantages and disadvantages of the co-culture model, identifies its technical bottlenecks, and proposes corresponding optimization strategies. By summarizing the latest research advancements in this co-culture model, our goal is to provide valuable insights for further model optimization and clinical application, thereby promoting immunological research and bridging the gap between experimental outcomes and clinical practice.
Collapse
Affiliation(s)
- Peng Li
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Minli Huang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yifan Ma
- Gansu University of traditional Chinese medicine, Lanzhou, 730030, Gansu, China
| | - Yongbin Zhang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
4
|
Meng S, Cao Y, Lu L, Li X, Sun S, Jiang F, Lu J, Fan D, Han X, Yao T. Quercetin Promote the Chemosensitivity in Organoids Derived from Patients with Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:993-1004. [PMID: 39720358 PMCID: PMC11668317 DOI: 10.2147/bctt.s494901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024]
Abstract
Aim The study aimed to culture organoids from tissues of patients with breast cancer (BC) and use the organoids to measure the sensitivity to quercetin and its combination with chemotherapeutic agents. Methods Four patient-derived organoids (PDOs) of BC were cultured. The proliferative activity and morphology of PDOs were evaluated on different generations and after resuscitation. H&E and immunohistochemical (IHC) staining were used to identify the pathological changes and the expression of biomarkers. The sensitivity to quercetin and chemotherapeutic agents and their combinations were evaluated using adenosine triphosphate (ATP) viability assays. Results We successfully obtained all PDOs from BC tissues. PDOs preserved their activity and morphology during generation passage. In addition, the pathological changes and expression patterns of estrogen receptor (ER), human epidermal growth factor receptor (HER2), and Ki67 of each PDO were consistent with their original tissues. All four PDOs were highly sensitive to quercetin, and their IC50 values were less than 22 μM. PDOs showed better sensitivity to docetaxel and epirubicin hydrochloride, but less sensitivity to cis-platinum. Combination with quercetin promoted the sensitivity to three chemotherapeutic agents. In particular, the IC50 value of cis-platinum greatly decreased. Conclusion We successfully established PDOs from patients with BC and demonstrated that quercetin can promote the sensitivity of chemotherapeutic agents in these PDOs.
Collapse
Affiliation(s)
- Shengwen Meng
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Yifan Cao
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Lei Lu
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Xuanhe Li
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Siyu Sun
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Fangqian Jiang
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Jianfei Lu
- Department of Breast and Thyroid Surgery, Bengbu First People’s Hospital, Bengbu, Anhui Province, People’s Republic of China
| | - Dongwei Fan
- Department of General Surgery, Affiliated Hospital of West Anhui Health Vocational College, Luan, Anhui Province, People’s Republic of China
| | - Xinxin Han
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| | - Tingjing Yao
- Department of Surgical Oncology, The Fourth Ward of Breast and Thyroid, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, People’s Republic of China
| |
Collapse
|
5
|
Zhao KY, Du YX, Cao HM, Su LY, Su XL, Li X. The biological macromolecules constructed Matrigel for cultured organoids in biomedical and tissue engineering. Colloids Surf B Biointerfaces 2024; 247:114435. [PMID: 39647422 DOI: 10.1016/j.colsurfb.2024.114435] [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: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Matrigel is the most commonly used matrix for 3D organoid cultures. Research on the biomaterial basis of Matrigel for organoid cultures is a highly challenging field. Currently, many studies focus on Matrigel-based biological macromolecules or combinations to construct natural Matrigel and synthetic hydrogel scaffolds based on collagen, peptides, polysaccharides, microbial transglutaminase, DNA supramolecules, and polymers for organoid culture. In this review, we discuss the limitations of both natural and synthetic Matrigel, and describe alternative scaffolds that have been employed for organoid cultures. The patient-derived organoids were constructed in different cancer types and limitations of animal-derived organoids based on the hydrogel or Matrigel. The constructed techniques utilizing 3D bioprinting platforms, air-liquid interface (ALI) culture, microfluidic culture, and organ-on-a-chip platform are summarized. Given the potential of organoids for a wide range of therapeutic, tissue engineering and pharmaceutical applications, it is indeed imperative to develop defined and customized hydrogels in addition to Matrigel.
Collapse
Affiliation(s)
- Ke-Yu Zhao
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Yi-Xiang Du
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Hui-Min Cao
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Li-Ya Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Xiu-Lan Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Xian Li
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
6
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
7
|
Fan G, Luo X, Shi Y, Wang Y, Ji L, Gong Y, Yang E, Chen C, Cui S, Ding H, Zhang Z, Wang J, Liu Y, Wang Z. FL118: A potential bladder cancer therapeutic compound targeting H2A.X identified through library screening. Bioorg Chem 2024; 153:107802. [PMID: 39244972 DOI: 10.1016/j.bioorg.2024.107802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
The treatment of bladder cancer is limited by low drug efficacy and drug resistance. Hence, this study aimed to screen and identify potential drug precursors and investigate their mechanism of action. A set of camptothecin derivatives showing high anti-tumor potential was selected from early-stage research or literature and synthesized to construct a compound library. A total of 135 compounds were screened in T24 and J82 cells, revealing that FL118 significantly inhibited the proliferation of GC (gemcitabine + cisplatin)-sensitive/insensitive cells. FL118 exhibited excellent penetration and killing ability in organoids and three GC-insensitive patient-derived xenografts. Chemical proteomic and docking calculations were employed to identify binding proteins, indicating that FL118 can bind into H2A.X and its entwined DNA. The results of Cellular thermal shift assay and surface plasmon resonance (Kd = 3.77E-6) support the above findings. Fluorescence localization revealed widespread binding of FL118 within the cell nucleus. Furthermore, WB showed that FL118 increased cellular DNA damage, resulting in significant cell cycle inhibition. The binding of FL118 to H2A.X hindered the damage repair process, leading to apoptosis. Controllable adverse reactions were observed in mice treated with FL118. In conclusion, FL118 may be a superior anti-bladder cancer compound that acts as a molecular glue binding to both H2A.X and DNA. The resistance mediated by the DNA damage repair to DNA damage caused by GC regimen can be reversed by FL118. This distinct mechanism of FL118 has the potential to complement existing mainstream treatment approaches for bladder cancer.
Collapse
Affiliation(s)
- Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Xiongfei Luo
- School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, Gansu Province, China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Yingru Wang
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Luhua Ji
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Engaung Yang
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Shu Cui
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Hui Ding
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Zhijun Zhang
- School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, Gansu Province, China
| | - Juan Wang
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China
| | - Yingqian Liu
- School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, Gansu Province, China.
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, Urinary System Disease Clinical Medical Research Center of Gansu Province, NO. 82 Cuiying Gate, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
8
|
Huang W, Xu Z, Li S, Zhou J, Zhao B. Living Biobanks of Organoids: Valuable Resource for Translational Research. Biopreserv Biobank 2024; 22:543-549. [PMID: 38959173 PMCID: PMC11656124 DOI: 10.1089/bio.2023.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
The emergence of organoids is considered a revolutionary model, changing the landscape of traditional translational research. These three-dimensional miniatures of human organs or tissues, cultivated from stem cells or biospecimens obtained from patients, faithfully replicate the structural and functional characteristics of specific target organs or tissues. In this extensive review, we explore the profound impact of organoids and assess the current state of living organoid biobanks, which are essential repositories for cryopreserving organoids derived from a variety of diseases. These resources hold significant value for translational research. We delve into the diverse origins of organoids, the underlying technologies, and their roles in recapitulating human development, disease modeling, as well as their potential applications in the pharmaceutical field. With a particular emphasis on biobanking organoids for prospective applications, we discuss how these advancements expedite the transition from bench to bedside translational research, thereby fostering personalized medicine and enriching our comprehension of human health.
Collapse
Affiliation(s)
- Wenqing Huang
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Zhaoting Xu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, Republic of China
| | - Shuang Li
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Junmei Zhou
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, Republic of China
| |
Collapse
|
9
|
Zhang X, Liu J, Ni Y, Yang Y, Tian T, Zheng X, Li Z, Huang R. Modeling Clinical Radioiodine Uptake By Using Organoids Derived From Differentiated Thyroid Cancer. Endocrinology 2024; 166:bqae162. [PMID: 39658331 DOI: 10.1210/endocr/bqae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024]
Abstract
Radioiodine-refractory differentiated thyroid cancer (RAI-R DTC) accounts for the vast majority of thyroid-related mortality and, until recently, there were limited preclinical models for iodine uptake prediction. In the current study, we aim to establish a primary tumor-derived organoid model of DTC and predict radioiodine (RAI) uptake of tumor residue. The genotypic and phenotypic features between organoid and parental tissue were compared. The RAI uptake assay was used to evaluate the organoid's RAI uptake capacity, and related patients' RAI whole-body scans were used to verify the assay's predictive sensitivity. A total of 20 patient-derived DTC organoids have been established. Whole-exome sequencing and immunofluorescence analysis demonstrated that organoids faithfully recapitulated main features of the original tumor tissue. RAI-avid organoids (n = 11) presented significantly higher RAI uptake than the RAI-refractory (RAI-R) group (n = 9; 384.4 ± 102.7 vs 54.2 ± 13.2 cpm/105 cells, P < .0001). A threshold value in organoids of less than 250 cpm/105 cell was found to have a predictive sensitivity of 95.0% for distinguishing RAI-R from RAI-avid patients when paired to clinical information. Notably, we found that several tyrosine kinase inhibitors moderately re-sensitize iodine uptake by using organoids derived from 3 patients with different genetic mutation backgrounds. In conclusion, patient-derived DTC organoids recapitulated the main characteristics of their parental tissues and preserved ability to uptake radioiodine, showing potential in the development of novel drugs to boost iodine avidity.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Jiaye Liu
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610000, China
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
- Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yinyun Ni
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ying Yang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Tian Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
- Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
| |
Collapse
|
10
|
Niazi V, Parseh B. Organoid models of breast cancer in precision medicine and translational research. Mol Biol Rep 2024; 52:2. [PMID: 39570495 DOI: 10.1007/s11033-024-10101-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
One of the most famous and heterogeneous cancers worldwide is breast cancer (BC). Owing to differences in the gene expression profiles and clinical features of distinct BC subtypes, different treatments are prescribed for patients. However, even with more thorough pathological evaluations of tumors than in the past, available treatments do not perform equally well for all individuals. Precision medicine is a new approach that considers the effects of patients' genes, lifestyle, and environment to choose the right treatment for an individual patient. As a powerful tool, the organoid culture system can maintain the morphological and genetic characteristics of patients' tumors. Evidence also shows that organoids have high predictive value for patient treatment. In this review, a variety of BC studies performed on organoid culture systems are evaluated. Additionally, the potential of using organoid models in BC translational research, especially in immunotherapy, drug screening, and precision medicine, has been reported.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Shastkola Street, Gorgan, 4918936316, Iran
| | - Benyamin Parseh
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran.
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Shastkola Street, Gorgan, 4918936316, Iran.
| |
Collapse
|
11
|
Melzer MK, Ma Y, Lindenmayer J, Morgenstern C, Wezel F, Zengerling F, Günes C, Gaisa NT, Kleger A, Bolenz C. Prospective pharmacotyping of urothelial carcinoma organoids for drug sensitivity prediction - feasibility and real world experience. Exp Hematol Oncol 2024; 13:112. [PMID: 39533373 PMCID: PMC11558855 DOI: 10.1186/s40164-024-00579-3] [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: 04/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Urothelial carcinoma (UC) of the urinary bladder has significant challenges in treatment due to its diverse genetic landscape and variable response to systemic therapy. In recent years, patient-derived organoids (PDOs) emerged as a novel tool to model primary tumors with higher resemblance than conventional 2D cell culture approaches. However, the potential of organoids to predict therapy response in a clinical setting remains to be evaluated. This study explores the clinical feasibility of PDOs for pharmacotyping in UC. Initially, we subjected tumor tissue specimens from 50 patients undergoing transurethral resection or radical cystectomy to organoid propagation, of whom 19 (38%) yielded PDOs suitable for drug sensitivity assessment. Notably, whole transcriptome-based analysis indicated that PDOs may show phenotypes distinct from their parental tumor tissue. Pharmacotyping within a clinically relevant timeframe [mean of 35.44 and 55 days for non-muscle invasive bladder cancer (NMIBC) and muscle invasive bladder cancer (MIBC), respectively] was achieved. Drug sensitivity analyses revealed marked differences between NMIBC and MIBC, with MIBC-derived organoids demonstrating higher chemosensitivity toward clinically relevant drugs. A case study correlating organoid response with patient treatment outcome illustrated the complexity of predicting chemotherapy efficacy, especially considering the rapid acquisition of drug resistance. We propose a workflow of prospective organoid-based pharmacotyping in UC, enabling further translational research and integration of this approach into clinical practice.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Department of Urology, Ulm University Hospital, Ulm, Germany.
- Institute for Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany.
| | - Yanchun Ma
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | | | - Clara Morgenstern
- Department of Urology, Ulm University Hospital, Ulm, Germany
- Institute for Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | | | - Cagatay Günes
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | | | - Alexander Kleger
- Institute for Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany.
- Core Facility Organoids, Ulm University, Ulm, Germany.
- Section for Interdisciplinary Pancreatology, Clinic for Internal Medicine I, Ulm University Hospital, Ulm, Germany.
| | | |
Collapse
|
12
|
Seven D, Baron S, Farin HF. Establishment of Patient-Derived Organoids from Colorectal Cancer Resection Samples. Methods Mol Biol 2024. [PMID: 39520596 DOI: 10.1007/7651_2024_575] [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: 11/16/2024]
Abstract
Colorectal cancer (CRC) organoids can serve as powerful preclinical cell models that accurately reflect individual tumor characteristics. Establishing a patient-derived CRC biobank facilitates a wide range of applications, including basic oncology research, new drug discovery, drug testing, and personalized medicine. This chapter details the process of generation of organoids from surgical samples of primary and metastatic CRC as well as from tumor adjacent normal colon tissues. Furthermore, best practices for cultivation and cryostorage of CRC organoids are described.
Collapse
Affiliation(s)
- Didem Seven
- Yeditepe University School of Medicine, Medical Genetics Department, Istanbul, Turkey.
| | - Szilvia Baron
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Tong L, Cui W, Zhang B, Fonseca P, Zhao Q, Zhang P, Xu B, Zhang Q, Li Z, Seashore-Ludlow B, Yang Y, Si L, Lundqvist A. Patient-derived organoids in precision cancer medicine. MED 2024; 5:1351-1377. [PMID: 39341206 DOI: 10.1016/j.medj.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024]
Abstract
Organoids are three-dimensional (3D) cultures, normally derived from stem cells, that replicate the complex structure and function of human tissues. They offer a physiologically relevant model to address important questions in cancer research. The generation of patient-derived organoids (PDOs) from various human cancers allows for deeper insights into tumor heterogeneity and spatial organization. Additionally, interrogating non-tumor stromal cells increases the relevance in studying the tumor microenvironment, thereby enhancing the relevance of PDOs in personalized medicine. PDOs mark a significant advancement in cancer research and patient care, signifying a shift toward more innovative and patient-centric approaches. This review covers aspects of PDO cultures to address the modeling of the tumor microenvironment, including extracellular matrices, air-liquid interface and microfluidic cultures, and organ-on-chip. Specifically, the role of PDOs as preclinical models in gene editing, molecular profiling, drug testing, and biomarker discovery and their potential for guiding personalized treatment in clinical practice are discussed.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Weiyingqi Cui
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Boya Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Pedro Fonseca
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Qian Zhao
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Ping Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Beibei Xu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qisi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Zeng G, Yu Y, Wang M, Liu J, He G, Yu S, Yan H, Yang L, Li H, Peng X. Advancing cancer research through organoid technology. J Transl Med 2024; 22:1007. [PMID: 39516934 PMCID: PMC11545094 DOI: 10.1186/s12967-024-05824-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
The complexity of tumors and the challenges associated with treatment often stem from the limitations of existing models in accurately replicating authentic tumors. Recently, organoid technology has emerged as an innovative platform for tumor research. This bioengineering approach enables researchers to simulate, in vitro, the interactions between tumors and their microenvironment, thereby enhancing the intricate interplay between tumor cells and their surroundings. Organoids also integrate multidimensional data, providing a novel paradigm for understanding tumor development and progression while facilitating precision therapy. Furthermore, advancements in imaging and genetic editing techniques have significantly augmented the potential of organoids in tumor research. This review explores the application of organoid technology for more precise tumor simulations and its specific contributions to cancer research advancements. Additionally, we discuss the challenges and evolving trends in developing comprehensive tumor models utilizing organoid technology.
Collapse
Affiliation(s)
- Guolong Zeng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yifan Yu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Meiting Wang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Sixuan Yu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Huining Yan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| |
Collapse
|
15
|
Monjaras-Avila CU, Luque-Badillo AC, Bacon JVM, Wyatt AW, So A, Chavez-Munoz C. A novel approach to engineering three-dimensional bladder tumor models for drug testing. Sci Rep 2024; 14:26883. [PMID: 39506094 PMCID: PMC11542063 DOI: 10.1038/s41598-024-78440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024] Open
Abstract
Bladder cancer (BCa) poses a significant health challenge, particularly affecting men with higher incidence and mortality rates. Addressing the need for improved predictive models in BCa treatment, this study introduces an innovative 3D in vitro patient-derived bladder cancer tumor model, utilizing decellularized pig bladders as scaffolds. Traditional 2D cell cultures, insufficient in replicating tumor microenvironments, have driven the development of sophisticated 3D models. The study successfully achieved pig bladder decellularization through multiple cycles of immersion in salt solutions, resulting in notable macroscopic and histological changes. This process confirmed the removal of cellular components while preserving the native extracellular matrix (ECM). Quantitative analysis demonstrated the efficacy of decellularization, with a remarkable reduction in DNA concentration, signifying the removal of over 95% of cellular material. In the development of the in vitro bladder cancer model, muscle invasive bladder cancer patients' cells were cultured within decellularized pig bladders, yielding a three-dimensional cancer model. Optimal results were attained using an air-liquid interface technique, with cells injected directly into the scaffold at three distinct time points. Histological evaluations showcased characteristics resembling in vivo tumors derived from bladder cancer patients' cells. To demonstrate the 3D cancer model's effectiveness as a drug screening platform, the study treated it with Cisplatin (Cis), Gemcitabine (Gem), and a combination of both drugs. Comprehensive cell viability assays and histological analyses illustrated changes in cell survival and proliferation. The model exhibited promising correlations with clinical outcomes, boasting an 83.3% reliability rate in predicting treatment responses. Comparison with traditional 2D cultures and spheroids underscored the 3D model's superiority in reliability, with an 83.3% predictive capacity compared to 50% for spheroids and 33.3% for 2D culture. Acknowledging limitations, such as the absence of immune and stromal components, the study suggests avenues for future improvements. In conclusion, this innovative 3D bladder cancer model, combining decellularization and patient-derived cells, marks a significant advancement in preclinical drug testing. Its potential for predicting treatment outcomes and capturing patient-specific responses opens new avenues for personalized medicine in bladder cancer therapeutics. Future refinements and validations with larger patient cohorts hold promise for revolutionizing BCa research and treatment strategies.
Collapse
Affiliation(s)
- C U Monjaras-Avila
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - A C Luque-Badillo
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - J V M Bacon
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - A W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - A So
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - C Chavez-Munoz
- Department of Medicine, Faculty of Medicine, University of British Columbia, H.B. 2660 Oak Street, Vancouver, BC, V6H3Z6, Canada.
| |
Collapse
|
16
|
Sase M, Sato T, Sato H, Miya F, Zhang S, Haeno H, Kajita M, Noguchi T, Mori Y, Ohteki T. Comparative analysis of tongue cancer organoids among patients identifies the heritable nature of minimal residual disease. Dev Cell 2024:S1534-5807(24)00607-5. [PMID: 39504967 DOI: 10.1016/j.devcel.2024.10.007] [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: 06/02/2024] [Revised: 08/13/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
The relapse of tongue cancer (TC) after chemotherapy is caused by minimal residual disease (MRD), which is a few remaining cancer cells after chemotherapy. To understand the mechanism of MRD in TC, we created a library of TC organoids (TCOs) from 28 untreated TC patients at diverse ages and cancer stages. These TCOs reproduced the primary TC tissues both in vitro and in a xenograft model, and several TCO lines survived after cisplatin treatment (chemo-resistant TCOs). Of note, the chemo-resistant TCOs showed "heritable" embryonic diapause-like features before treatment and activation of the autophagy and cholesterol biosynthetic pathways. Importantly, inhibiting these pathways with specific inhibitors converted the chemo-resistant TCOs into chemo-sensitive TCOs. Conversely, autophagy activation with mTOR inhibitors conferred chemo-resistance on the chemo-sensitive TCOs. This unique model provides insights into the mechanism of MRD formation in TCs, leading to effective therapeutic approaches to reduce the recurrence of TC.
Collapse
Affiliation(s)
- Miwako Sase
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University [TMDU]), Tokyo 113-8510, Japan; Department of Dentistry, Oral, and Maxillofacial Surgery, Jichi Medical University, Tochigi 329-0498, Japan
| | - Taku Sato
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University [TMDU]), Tokyo 113-8510, Japan; Department of Biochemistry and Molecular Biology, Nippon Medical School Graduate School of Medicine, Tokyo 113-8603, Japan
| | - Hajime Sato
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University [TMDU]), Tokyo 113-8510, Japan; Department of Dentistry, Oral, and Maxillofacial Surgery, Jichi Medical University, Tochigi 329-0498, Japan
| | - Fuyuki Miya
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; Center for Medical Genetics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shicheng Zhang
- Research Institute for Biomedical Science, Tokyo University of Science, Chiba 278-0022, Japan
| | - Hiroshi Haeno
- Research Institute for Biomedical Science, Tokyo University of Science, Chiba 278-0022, Japan
| | - Mihoko Kajita
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University [TMDU]), Tokyo 113-8510, Japan
| | - Tadahide Noguchi
- Department of Dentistry, Oral, and Maxillofacial Surgery, Jichi Medical University, Tochigi 329-0498, Japan
| | - Yoshiyuki Mori
- Department of Dentistry, Oral, and Maxillofacial Surgery, Jichi Medical University, Tochigi 329-0498, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University [TMDU]), Tokyo 113-8510, Japan.
| |
Collapse
|
17
|
Merrill NM, Kaffenberger SD, Bao L, Vandecan N, Goo L, Apfel A, Cheng X, Qin Z, Liu CJ, Bankhead A, Wang Y, Kathawate V, Tudrick L, Serhan HA, Farah Z, Ellimoottil C, Hafez KS, Herrel LA, Montgomery JS, Morgan TM, Salami SS, Weizer AZ, Ulintz PJ, Day ML, Soellner MB, Palmbos PL, Merajver SD, Udager AM. Integrative Drug Screening and Multiomic Characterization of Patient-derived Bladder Cancer Organoids Reveal Novel Molecular Correlates of Gemcitabine Response. Eur Urol 2024; 86:434-444. [PMID: 39155193 DOI: 10.1016/j.eururo.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Predicting response to therapy for each patient's tumor is critical to improving long-term outcomes for muscle-invasive bladder cancer. This study aims to establish ex vivo bladder cancer patient-derived organoid (PDO) models that are representative of patients' tumors and determine the potential efficacy of standard of care and curated experimental therapies. METHODS Tumor material was collected prospectively from consented bladder cancer patients to generate short-term PDO models, which were screened against a panel of clinically relevant drugs in ex vivo three-dimensional culture. Multiomic profiling was utilized to validate the PDO models, establish the molecular characteristics of each tumor, and identify potential biomarkers of drug response. Gene expression (GEX) patterns between paired primary tissue and PDO samples were assessed using Spearman's rank correlation coefficients. Molecular correlates of therapy response were identified using Pearson correlation coefficients and Kruskal-Wallis tests with Dunn's post hoc pairwise comparison testing. KEY FINDINGS AND LIMITATIONS A total of 106 tumors were collected from 97 patients, with 65 samples yielding sufficient material for complete multiomic molecular characterization and PDO screening with six to 32 drugs/combinations. Short-term PDOs faithfully represent the tumor molecular characteristics, maintain diverse cell types, and avoid shifts in GEX-based subtyping that accompany long-term PDO cultures. Utilizing an integrative approach, novel correlations between ex vivo drug responses and genomic alterations, GEX, and protein expression were identified, including a multiomic signature of gemcitabine response. The positive predictive value of ex vivo drug responses and the novel multiomic gemcitabine response signature need to be validated in future studies. CONCLUSIONS AND CLINICAL IMPLICATIONS Short-term PDO cultures retain the molecular characteristics of tumor tissue and avoid shifts in expression-based subtyping that have plagued long-term cultures. Integration of multiomic profiling and ex vivo drug screening data identifies potential predictive biomarkers, including a novel signature of gemcitabine response. PATIENT SUMMARY Better models are needed to predict patient response to therapy in bladder cancer. We developed a platform that uses short-term culture to best mimic each patient's tumor and assess potential sensitivity to therapeutics.
Collapse
Affiliation(s)
- Nathan M Merrill
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Liwei Bao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Laura Goo
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Athena Apfel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xu Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhaoping Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chia-Jen Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Armand Bankhead
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yin Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Varun Kathawate
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lila Tudrick
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Habib A Serhan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zackariah Farah
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chad Ellimoottil
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Khaled S Hafez
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Lindsey A Herrel
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey S Montgomery
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Todd M Morgan
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Simpa S Salami
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Alon Z Weizer
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Peter J Ulintz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark L Day
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | | | - Phillip L Palmbos
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sofia D Merajver
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Aaron M Udager
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Guo Z, Li Z, Wang J, Jiang H, Wang X, Sun Y, Huang W. Modeling bladder cancer in the laboratory: Insights from patient-derived organoids. Biochim Biophys Acta Rev Cancer 2024; 1879:189199. [PMID: 39419296 DOI: 10.1016/j.bbcan.2024.189199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Bladder cancer (BCa) is the most common malignant tumor of the urinary system. Current treatments often have poor efficacy and carry a high risk of recurrence and progression due to the lack of consideration of tumor heterogeneity. Patient-derived organoids (PDOs) are three-dimensional tissue cultures that preserve tumor heterogeneity and clinical relevance better than cancer cell lines. Moreover, PDOs are more cost-effective and efficient to cultivate compared to patient-derived tumor xenografts, while closely mirroring the tissue and genetic characteristics of their source tissues. The development of PDOs involves critical steps such as sample selection and processing, culture medium optimization, matrix selection, and improvements in culture methods. This review summarizes the methodologies for generating PDOs from patients with BCa and discusses the current advancements in drug sensitivity testing, immunotherapy, living biobanks, drug screening, and mechanistic studies, highlighting their role in advancing personalized medicine.
Collapse
Affiliation(s)
- Zikai Guo
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China
| | - Jia Wang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China
| | - Hongxiao Jiang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China; Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xu Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei 230061, China
| | - Yangyang Sun
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China; Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Weiren Huang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center of Shenzhen University, Shenzhen 518039, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China; Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
19
|
Liu X, Han X, Wei S, Zhang C. Case report: Patient-derived organoids promoting personalized treatment in invasive urothelial carcinoma. Front Oncol 2024; 14:1424677. [PMID: 39555454 PMCID: PMC11563985 DOI: 10.3389/fonc.2024.1424677] [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: 04/28/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024] Open
Abstract
Tumor organoids, an in-vitro three-dimensional model, possess high potential for investigating tumor biology and treatment response and have been demonstrated more appropriate for drug assessment than two-dimensional cultures. Herein, we described two cases of invasive high-grade urothelial carcinoma who underwent radical cystectomy successfully following use of patient-derived organoids (PDOs) for drug screening to inform therapeutic decisions. In these two cases, the PDOs cultured by biopsy tissues were both sensitive to the combination of gemcitabine and cisplatin. After neoadjuvant chemotherapy (NAC) with gemcitabine and cisplatin, the patients responded well, and radical cystectomy was performed successfully. No recurrence or metastasis was observed within 6 months after surgery. This small case series suggests that the patient-derived urothelial carcinoma organoids contribute to optimizing NAC options to guide personalized treatment and improve the survival outcomes.
Collapse
Affiliation(s)
- Xun Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xuebing Han
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Shuqing Wei
- Department of General Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Changwen Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
20
|
Plage H, Dressler FF, Fendler A. Patient-derived Models: A Promising Frontier in Testing the Efficacy of Drugs for Bladder Cancer. Eur Urol 2024; 86:445-446. [PMID: 39183091 DOI: 10.1016/j.eururo.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Affiliation(s)
- Henning Plage
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Franz F Dressler
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Fendler
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
21
|
Abbasian MH, Sobhani N, Sisakht MM, D'Angelo A, Sirico M, Roudi R. Patient-Derived Organoids: A Game-Changer in Personalized Cancer Medicine. Stem Cell Rev Rep 2024:10.1007/s12015-024-10805-4. [PMID: 39432173 DOI: 10.1007/s12015-024-10805-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Research on cancer therapies has benefited from predictive tools capable of simulating treatment response and other disease characteristics in a personalized manner, in particular three-dimensional cell culture models. Such models include tumor-derived spheroids, multicellular spheroids including organotypic multicellular spheroids, and tumor-derived organoids. Additionally, organoids can be grown from various cancer cell types, such as pluripotent stem cells and induced pluripotent stem cells, progenitor cells, and adult stem cells. Although patient-derived xenografts and genetically engineered mouse models replicate human disease in vivo, organoids are less expensive, less labor intensive, and less time-consuming, all-important aspects in high-throughput settings. Like in vivo models, organoids mimic the three-dimensional structure, cellular heterogeneity, and functions of primary tissues, with the advantage of representing the normal oxygen conditions of patient organs. In this review, we summarize the use of organoids in disease modeling, drug discovery, toxicity testing, and precision oncology. We also summarize the current clinical trials using organoids.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Mahsa Mollapour Sisakht
- Faculty of Pharmacy, Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AX, UK
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 PMCID: PMC11573787 DOI: 10.1016/j.crmeth.2024.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Liu Y, Lankadasari M, Rosiene J, Johnson KE, Zhou J, Bapat S, Chow-Tsang LFL, Tian H, Mastrogiacomo B, He D, Connolly JG, Lengel HB, Caso R, Dunne EG, Fick CN, Rocco G, Sihag S, Isbell JM, Bott MJ, Li BT, Lito P, Brennan CW, Bilsky MH, Rekhtman N, Adusumilli PS, Mayo MW, Imielinski M, Jones DR. Modeling lung adenocarcinoma metastases using patient-derived organoids. Cell Rep Med 2024; 5:101777. [PMID: 39413736 PMCID: PMC11513837 DOI: 10.1016/j.xcrm.2024.101777] [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: 02/13/2024] [Revised: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
Approximately 50% of patients with surgically resected early-stage lung cancer develop distant metastasis. At present, there is no in vivo metastasis model to investigate the biology of human lung cancer metastases. Using well-characterized lung adenocarcinoma (LUAD) patient-derived organoids (PDOs), we establish an in vivo metastasis model that preserves the biologic features of human metastases. Results of whole-genome and RNA sequencing establish that our in vivo PDO metastasis model can be used to study clonality and tumor evolution and to identify biomarkers related to organotropism. Investigation of the response of KRASG12C PDOs to sotorasib demonstrates that the model can examine the efficacy of treatments to suppress metastasis and identify mechanisms of drug resistance. Finally, our PDO model cocultured with autologous peripheral blood mononuclear cells can potentially be used to determine the optimal immune-priming strategy for individual patients with LUAD.
Collapse
Affiliation(s)
- Yuan Liu
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manendra Lankadasari
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joel Rosiene
- Department of Pathology, New York University, New York, NY, USA
| | - Kofi E Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Tri-Institutional PhD Program in Computational Biology and Medicine, New York, NY, USA
| | - Juan Zhou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samhita Bapat
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lai-Fong L Chow-Tsang
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Huasong Tian
- Department of Pathology, New York University, New York, NY, USA
| | - Brooke Mastrogiacomo
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Di He
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James G Connolly
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harry B Lengel
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raul Caso
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth G Dunne
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cameron N Fick
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gaetano Rocco
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James M Isbell
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mathew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bob T Li
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Piro Lito
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cameron W Brennan
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark H Bilsky
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marty W Mayo
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | | | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, Tan Y, Liu Z. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater 2024:e2401843. [PMID: 39397335 DOI: 10.1002/adhm.202401843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/25/2024] [Indexed: 10/15/2024]
Abstract
The development of personalized precision medicine has become a pivotal focus in modern healthcare. Organoids-on-a-Chip (OoCs), a groundbreaking fusion of organoid culture and microfluidic chip technology, has emerged as a promising approach to advancing patient-specific treatment strategies. In this review, the diverse applications of OoCs are explored, particularly their pivotal role in personalized precision medicine, and their potential as a cutting-edge technology is highlighted. By utilizing patient-derived organoids, OoCs offer a pathway to optimize treatments, create precise disease models, investigate disease mechanisms, conduct drug screenings, and individualize therapeutic strategies. The emphasis is on the significance of this technological fusion in revolutionizing healthcare and improving patient outcomes. Furthermore, the transformative potential of personalized precision medicine, future prospects, and ongoing advancements in the field, with a focus on genomic medicine, multi-omics integration, and ethical frameworks are discussed. The convergence of these innovations can empower patients, redefine treatment approaches, and shape the future of healthcare.
Collapse
Affiliation(s)
- Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
25
|
Zhu Q, He X, Liu J, Wang H, Shan X, Song G, Zhang L, Zhao Y, Yin X. DNA origami assembled spheroid for evaluating cytotoxicity and infiltration of chimeric antigen receptor macrophage (CAR-M). Commun Biol 2024; 7:1302. [PMID: 39390143 PMCID: PMC11467189 DOI: 10.1038/s42003-024-07009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have shown remarkable results in patients with hematological malignancies. However, their success in treating solid tumors has been limited. As an alternative candidate for the CAR therapy, CAR-macrophages (CAR-M) have demonstrated activation and phagocytosis directed by tumor-associated antigen (TAA), showing promise in the treatment of solid tumors. Nevertheless, the mechanisms by which CARs direct tumor chemotaxis and invasion of CAR-M remain poorly understood. In this study, we aim to investigate the role of CARs in CAR-M attachment and infiltration using 3D tumor spheroids, which were created by utilizing a novel self-assembling nucleic acid nanostructure decorated living cells (NAC). Our results demonstrated that CAR-M exhibited higher invasion and killing capacity in 2D model and 3D tumor spheroids. In summary, the 3D NAC assembled tumor spheroid model provides a suitable platform for target screening and pharmacodynamic evaluation of CAR-M.
Collapse
Affiliation(s)
- Qinyao Zhu
- RocRock Biotechnology Co. Ltd, Suzhou, China
| | - Xiaofang He
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology Co., Ltd, Suzhou, China
| | - Junhua Liu
- RocRock Biotechnology Co. Ltd, Suzhou, China
| | - Heming Wang
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology Co., Ltd, Suzhou, China
| | | | - Guangqi Song
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology Co., Ltd, Suzhou, China.
| | - Luo Zhang
- Research Center of Bioengineering, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.
| | - Yicheng Zhao
- China-Japan Union Hospital of Jilin University, Changchun, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Xiushan Yin
- RocRock Biotechnology Co. Ltd, Suzhou, China.
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Biomedical Sciences College, Shandong First Medical University, Jinan, China.
| |
Collapse
|
26
|
Vasiliadou I, Cattaneo C, Chan PYK, Henley-Smith R, Gregson-Williams H, Collins L, Wojewodka G, Guerrero-Urbano T, Jeannon JP, Connor S, Davis J, Pasto A, Mustapha R, Ng T, Kong A. Correlation of the treatment sensitivity of patient-derived organoids with treatment outcomes in patients with head and neck cancer (SOTO): protocol for a prospective observational study. BMJ Open 2024; 14:e084176. [PMID: 39389599 PMCID: PMC11474813 DOI: 10.1136/bmjopen-2024-084176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Organoids have been successfully used in several areas of cancer research and large living biobanks of patient-derived organoids (PDOs) have been developed from various malignancies. The characteristics of the original tumour tissue such as mutation signatures, phenotype and genetic diversity are well preserved in organoids, thus showing promising results for the use of this model in translational research. In this study, we aim to assess whether we can generate PDOs from head and neck squamous cell carcinoma (HNSCC) samples and whether PDOs can be used to predict treatment sensitivity in HNSCC patients as well as to explore potential biomarkers. METHODS AND ANALYSIS This is a prospective observational study at a single centre (Guy's and St Thomas' NHS Foundation Trust) to generate PDOs from patients' samples to assess treatment response and to correlate with patients' treatment outcomes. Patients will be included if they are diagnosed with HNSCC undergoing curative treatment (primary surgery or radiotherapy) or presenting with recurrent or metastatic cancers and they will be categorised into three groups (cohort 1: primary surgery, cohort 2: primary radiotherapy and cohort 3: recurrent/metastatic disease). Research tumour samples will be collected and processed into PDOs and chemosensitivity/radiosensitivity will be assessed using established methods. Moreover, blood and other biological samples (eg, saliva) will be collected at different time intervals during treatment and will be processed in the laboratory for plasma and peripheral blood mononuclear cell (PBMC) isolation. Plasma and saliva will be used for circulating tumour DNA analysis and PBMC will be stored for assessment of the peripheral immune characteristics of the patients as well as to perform co-culture experiments with PDOs. SOTO study (correlation of the treatment Sensitivity of patient-derived Organoids with Treatment Outcomes in patients with head and neck cancer) uses the collaboration of several specialties in head and neck cancer and has the potential to explore multiple areas of research with the aim of offering a valid and effective approach to personalised medicine for cancer patients. ETHICS AND DISSEMINATION This study was approved by North West-Greater Manchester South Research Ethics Committee (REC Ref: 22/NW/0023) on 21 March 2022. An informed consent will be obtained from all participants prior to inclusion in the study. Results will be disseminated via peer-reviewed publications and presentations at international conferences. TRIAL REGISTRATION NUMBER NCT05400239.
Collapse
Affiliation(s)
| | | | | | - Rhonda Henley-Smith
- Head and Neck Pathology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | | | - Lisette Collins
- Head and Neck Pathology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | | | | | | | - Steve Connor
- Head and Neck Radiology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | - Jessica Davis
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Anna Pasto
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Rami Mustapha
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Tony Ng
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Anthony Kong
- Comprehensive Cancer Centre, King's College London, London, UK
| |
Collapse
|
27
|
Zhang Q, Zhang M. Recent advances in lung cancer organoid (tumoroid) research (Review). Exp Ther Med 2024; 28:383. [PMID: 39161616 PMCID: PMC11332118 DOI: 10.3892/etm.2024.12672] [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: 07/13/2023] [Accepted: 02/01/2024] [Indexed: 08/21/2024] Open
Abstract
Lung cancer is the most critical type of malignant tumor that threatens human health. Traditional preclinical models have certain defects; for example, they cannot accurately reflect the characteristics of lung cancer and their development is costly and time-consuming. Through self-organization, cancer stem cells (CSCs) generate cancer organoids that have a structure similar to that of lung cancer tissues, overcoming to some extent the aforementioned challenges, thus enabling them to have broader application prospects. Lung cancer organoid (LCO) development methods can be divided into three broad categories based on the source of cells, which include cell lines, patient-derived xenografts and patient tumor tissue/pleural effusion. There are 17 different methods that have been described for the development of LCOs. These methods can be further merged into six categories based on the source of cells, the pre-treatment method used, the composition of the medium and the culture scaffold. These categories are: i) CSCs induced by defined transcription factors; ii) suspension culture; iii) relative optimal culture medium; iv) suboptimal culture medium; v) mechanical digestion and suboptimal culture medium; and vi) hydrogel scaffold. In the current review, the advantages and disadvantages of each of the aforementioned methods are summarized, and references for supporting studies are cited.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Clinical Laboratory, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Mingyang Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
28
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
29
|
Qin X, Tape CJ. Functional analysis of cell plasticity using single-cell technologies. Trends Cell Biol 2024; 34:854-864. [PMID: 38355348 DOI: 10.1016/j.tcb.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Metazoan organisms are heterocellular systems composed of hundreds of different cell types, which arise from an isogenic genome through differentiation. Cellular 'plasticity' further enables cells to alter their fate in response to exogenous cues and is involved in a variety of processes, such as wound healing, infection, and cancer. Recent advances in cellular model systems, high-dimensional single-cell technologies, and lineage tracing have sparked a renaissance in plasticity research. Here, we discuss the definition of cell plasticity, evaluate state-of-the-art model systems and techniques to study cell-fate dynamics, and explore the application of single-cell technologies to obtain functional insights into cell plasticity in healthy and diseased tissues. The integration of advanced biomimetic model systems, single-cell technologies, and high-throughput perturbation studies is enabling a new era of research into non-genetic plasticity in metazoan systems.
Collapse
Affiliation(s)
- Xiao Qin
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK.
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
30
|
Esposito A, Ferraresi A, Vallino L, Garavaglia B, Dhanasekaran DN, Isidoro C. Three-Dimensional In Vitro Cell Cultures as a Feasible and Promising Alternative to Two-Dimensional and Animal Models in Cancer Research. Int J Biol Sci 2024; 20:5293-5311. [PMID: 39430243 PMCID: PMC11488579 DOI: 10.7150/ijbs.96469] [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: 03/20/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer represents one of the diseases with the highest mortality rate worldwide. The burden of cancer continues to increase, not only affecting the health-related quality of life of patients but also causing an elevated global financial impact. The complexity and heterogeneity of cancer pose significant challenges in research and clinical practice, contributing to increase the failure rate of clinical trials for antitumoral drugs. This is partially due to the fact that preclinical models still present important limitations in faithfully recapitulating human tumors to serve as reliable indicators of drug effectiveness. Up to now, research and development strategies employ expensive animal models (including the so-called "humanized mice") that not only raise ethical concerns, but also frequently fail to accurately predict responses to anticancer drugs because they do not faithfully replicate human physiology as well as the patient's tumor microenvironment. On the other side, traditional two-dimensional (2D) cell cultures fail to adequately reproduce the structural organization of tumor and the cellular heterogeneity found in vivo. The growing necessity to develop more accurate cancer models has increasingly emphasized the importance of three-dimensional (3D) in vitro cell cultures, such as cancer-derived spheroids and organoids, as promising alternatives to bridge the gap between 2D and animal models. In this review, we provide a brief overview focusing on 3D in vitro cell cultures as preclinical models capable of properly reproducing the tissue organization, biological composition, and complexity of in vivo tumors in a fine-tuned microenvironment. Despite their limitations, these models collectively enhance our understanding of the mechanisms underlying cancer and may offer the potential for a more reliable assessment of drug efficacy before clinical testing and, consequently, improve therapeutic outcomes for cancer patients.
Collapse
Affiliation(s)
- Andrea Esposito
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Beatrice Garavaglia
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| |
Collapse
|
31
|
Jiang M, Tu R, Pan Y, Cui Y, Qi X, Qin H, Liu L, Wang X, Xue Y, Xu Y, Peng Z, Zhang C, Yang J. Patient-derived organoids and mini-PDX for predicting MET N375S-mutated lung cancer patient clinical therapeutic response. Heliyon 2024; 10:e37884. [PMID: 39328538 PMCID: PMC11425092 DOI: 10.1016/j.heliyon.2024.e37884] [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: 07/02/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Lung cancer as a molecularly and histologically high heterogonous disease, there is an urgent need to predict lung cancer patients' responses to anti-cancer treatment, and patient-derived organoids (PDOs) have been recognized as a valuable platform for preclinical drug screening. In this study, we successfully established 26 PDO lines from various subtypes of lung cancers including benign tumor, adenocarcinoma, squamous cell carcinoma, adenosquamous carcinoma, large-cell carcinoma, and small-cell carcinoma. These PDOs were shown to retain the major genomic and histological characteristics of primary tumors and remain stable during long-term culture. With the help of targeted genomic sequencing, we found that lung cancer that harbors METN375S mutation is selectively sensitive to afatinib, and a combination of afatinib and gemcitabine induced synthetic lethality in PDO and mini-PDX models. In summary, our findings demonstrate the potential of PDO in predicting lung cancer drug response, and reveal a promising strategy for METN375S mutant lung cancer treatment.
Collapse
Affiliation(s)
- Meng Jiang
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Rongfu Tu
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Cancer Precision Medicine, The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Yiwen Pan
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yuxin Cui
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xin Qi
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Hongyu Qin
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Lijuan Liu
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xiaorui Wang
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Ying Xue
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yao Xu
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Ziyang Peng
- Department of Thoracic Surgery, Department of Thoracic Surgery and Oncology, Cancer Center, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710061, China
| | - Chengsheng Zhang
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Yang
- Precision Medicine Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
32
|
Tuo Z, Gao M, Jiang C, Zhang D, Chen X, Jiang Z, Wang J. Construction of M2 macrophage-related gene signature for predicting prognosis and revealing different immunotherapy response in bladder cancer patients. Clin Transl Oncol 2024:10.1007/s12094-024-03698-9. [PMID: 39347941 DOI: 10.1007/s12094-024-03698-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Bladder cancer development is closely associated with the dynamic interaction and communication between M2 macrophages and tumor cells. However, specific biomarkers for targeting M2 macrophages in immunotherapy remain limited and require further investigation. METHODS In this study, we identified key co-expressed genes in M2 macrophages and developed gene signatures to predict prognosis and immunotherapy response in patients. Public database provided the bioinformatics data used in the analysis. We created and verified an M2 macrophage-related gene signature in these datasets using Lasso-Cox analysis. RESULTS The predictive value and immunological functions of our risk model were examined in bladder cancer patients, and 158 genes were found to be significantly positively correlated with M2 macrophages. Moreover, we identified two molecular subgroups of bladder cancer with markedly different immunological profiles and clinical prognoses. The five key risk genes identified in this model were validated, including CALU, ECM1, LRP1, CYTL1, and CCDC102B, demonstrating the model can accurately predict prognosis and identify unique responses to immunotherapy in patients with bladder cancer. CONCLUSIONS In summary, we constructed and validated a five-gene signature related to M2 macrophages, which shows strong potential for forecasting bladder cancer prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Zhouting Tuo
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Mingzhu Gao
- Department of Oncology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Chao Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Duobing Zhang
- Department of Urology, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, China
- Department of Urology, Suzhou Municipal Hospital of Anhui Province, Suzhou, 234000, China
| | - Xin Chen
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhiwei Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jinyou Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
33
|
Glibetic N, Bowman S, Skaggs T, Weichhaus M. The Use of Patient-Derived Organoids in the Study of Molecular Metabolic Adaptation in Breast Cancer. Int J Mol Sci 2024; 25:10503. [PMID: 39408832 PMCID: PMC11477048 DOI: 10.3390/ijms251910503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Around 13% of women will likely develop breast cancer during their lifetime. Advances in cancer metabolism research have identified a range of metabolic reprogramming events, such as altered glucose and amino acid uptake, increased reliance on glycolysis, and interactions with the tumor microenvironment (TME), all of which present new opportunities for targeted therapies. However, studying these metabolic networks is challenging in traditional 2D cell cultures, which often fail to replicate the three-dimensional architecture and dynamic interactions of real tumors. To address this, organoid models have emerged as powerful tools. Tumor organoids are 3D cultures, often derived from patient tissue, that more accurately mimic the structural and functional properties of actual tumor tissues in vivo, offering a more realistic model for investigating cancer metabolism. This review explores the unique metabolic adaptations of breast cancer and discusses how organoid models can provide deeper insights into these processes. We evaluate the most advanced tools for studying cancer metabolism in three-dimensional culture models, including optical metabolic imaging (OMI), matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), and recent advances in conventional techniques applied to 3D cultures. Finally, we explore the progress made in identifying and targeting potential therapeutic targets in breast cancer metabolism.
Collapse
Affiliation(s)
- Natalija Glibetic
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- The IDeA Networks of Biomedical Research Excellence (INBRE) Program, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
- United Nations CIFAL Honolulu Center, Chaminade University, Honolulu, HI 96816, USA
| | - Scott Bowman
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biochemistry, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Tia Skaggs
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biology, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Michael Weichhaus
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
| |
Collapse
|
34
|
Zhou LF, Liao HY, Han Y, Zhao Y. The use of organoids in creating immune microenvironments and treating gynecological tumors. J Transl Med 2024; 22:856. [PMID: 39313812 PMCID: PMC11421176 DOI: 10.1186/s12967-024-05649-y] [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: 07/27/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Owing to patient-derived tumor tissues and cells, significant advances have been made in personalized cancer treatment and precision medicine, with cancer stem cell-derived three-dimensional tumor organoids serving as crucial in vitro models that accurately replicate the structural, phenotypic, and genetic characteristics of tumors. However, despite their extensive use in drug testing, genome editing, and transplantation for facilitating personalized treatment approaches in clinical practice, the inadequate capacity of these organoids to effectively model immune cells and stromal components within the tumor microenvironment limits their potential. Additionally, effective clinical immunotherapy has led the tumor immune microenvironment to garner considerable attention, increasing the demand for simulating patient-specific tumor-immune interactions. Consequently, co-culture techniques integrating tumor organoids with immune cells and tumor microenvironment constituents have been developed to expand the possibilities for personalized drug response investigations, with recent advancements enhancing the understanding of the strengths, limitations, and applicability of the co-culture approach. Herein, the recent advancements in the field of tumor organoids have been comprehensively reviewed, specifically highlighting the tumor organoid co-culture-related developments with various immune cell models and their implications for clinical research. Furthermore, this review delineates the current state of research and application of organoid models regarding the therapeutic approaches and related challenges for gynecological tumors. This study may provide a theoretical basis for further research on the use of patient-derived organoids in tumor immunity, drug development, and precision medicine.
Collapse
Affiliation(s)
- Ling-Feng Zhou
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Hui-Yan Liao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Yang Han
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China.
| |
Collapse
|
35
|
He L, Wang L, Yu X, Tang Y, Jiang Z, Yang G, Liu Z, Li W. Full-course NIR-II imaging-navigated fractionated photodynamic therapy of bladder tumours with X-ray-activated nanotransducers. Nat Commun 2024; 15:8240. [PMID: 39300124 DOI: 10.1038/s41467-024-52607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
The poor 5-year survival rate for bladder cancers is associated with the lack of efficient diagnostic and treatment techniques. Despite cystoscopy-assisted photomedicine and external radiation being promising modalities to supplement or replace surgery, they remain invasive or fail to provide real-time navigation. Here, we report non-invasive fractionated photodynamic therapy of bladder cancer with full-course real-time near-infrared-II imaging based on engineered X-ray-activated nanotransducers that contain lanthanide-doped nanoscintillators with concurrent emissions in visible and the second near-infrared regions and conjugated photosensitizers. Following intravesical instillation in mice with carcinogen-induced autochthonous bladder tumours, tumour-homing peptide-labelled nanotransducers realize enhanced tumour regression, robust recurrence inhibition, improved survival rates, and restored immune homeostasis under X-ray irradiation with accompanied near-infrared-II imaging. On-demand fractionated photodynamic therapy with customized doses is further achieved based on quantifiable near-infrared-II imaging signal-to-background ratios. Our study presents a promising non-invasive strategy to confront the current bladder cancer dilemma from diagnosis to treatment and prognosis.
Collapse
Affiliation(s)
- Liangrui He
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, PR China
| | - Liyang Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xujiang Yu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Yizhang Tang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zhao Jiang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, PR China
| | - Guoliang Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, PR China.
| | - Wanwan Li
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
36
|
Dinić J, Jovanović Stojanov S, Dragoj M, Grozdanić M, Podolski-Renić A, Pešić M. Cancer Patient-Derived Cell-Based Models: Applications and Challenges in Functional Precision Medicine. Life (Basel) 2024; 14:1142. [PMID: 39337925 PMCID: PMC11433531 DOI: 10.3390/life14091142] [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: 07/31/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The field of oncology has witnessed remarkable progress in personalized cancer therapy. Functional precision medicine has emerged as a promising avenue for achieving superior treatment outcomes by integrating omics profiling and sensitivity testing of patient-derived cancer cells. This review paper provides an in-depth analysis of the evolution of cancer-directed drugs, resistance mechanisms, and the role of functional precision medicine platforms in revolutionizing individualized treatment strategies. Using two-dimensional (2D) and three-dimensional (3D) cell cultures, patient-derived xenograft (PDX) models, and advanced functional assays has significantly improved our understanding of tumor behavior and drug response. This progress will lead to identifying more effective treatments for more patients. Considering the limited eligibility of patients based on a genome-targeted approach for receiving targeted therapy, functional precision medicine provides unprecedented opportunities for customizing medical interventions according to individual patient traits and individual drug responses. This review delineates the current landscape, explores limitations, and presents future perspectives to inspire ongoing advancements in functional precision medicine for personalized cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (S.J.S.); (M.D.); (M.G.); (A.P.-R.)
| |
Collapse
|
37
|
Radić M, Egger M, Kruithof-de Julio M, Seiler R. Patient-derived Organoids in Bladder Cancer: Opportunities and Challenges. Eur Urol Focus 2024:S2405-4569(24)00165-2. [PMID: 39232905 DOI: 10.1016/j.euf.2024.08.008] [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: 05/24/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND AND OBJECTIVE Bladder cancer (BLCa) remains a prevalent malignancy with high recurrence rates and limited treatment options. In recent years, patient-derived organoids (PDOs) have emerged as a promising platform for studying cancer biology and therapeutic responses in a personalized manner. Using drug screening, PDOs facilitate the identification of novel therapeutic agents and translational treatment strategies. Moreover, their ability to model patient-specific responses to treatments holds promise for predicting clinical outcomes and guiding treatment decisions. This exploratory review aims to investigate the potential of PDOs in advancing BLCa research and treatment, with an emphasis on translational clinical approaches. Furthermore, we analyze the feasibility of deriving PDOs from minimally invasive blood and urine samples. METHODS In addition to exploring hypothetical applications of PDOs for predicting patient outcomes and their ability to model different stages of BLCa, we conducted a comprehensive PubMed search on already published data as well as comprehensive screening of currently ongoing trials implementing PDOs in precision medicine in cancer patients irrespective of the tumor entity. KEY FINDINGS AND LIMITATIONS While the research on BLCa PDOs is advancing rapidly, data on both BLCa PDO research and their clinical application are scarce. Owing to this fact, a narrative review format was chosen for this publication. CONCLUSIONS AND CLINICAL IMPLICATIONS BLCa PDOs have the potential to influence the domain of precision medicine and enhance personalized cancer treatment strategies. However, standardized protocols for PDO generation, their ideal clinical application, as well as their impact on outcomes remain to be determined. PATIENT SUMMARY In this review, we discuss the current state and future needs for the use of patient-derived organoids, small three-dimensional avatars of tumor cells, in bladder cancer. Patient-derived bladder cancer organoids offer a more personalized approach to studying and treating bladder cancer, providing a model that closely resembles the patient's own tumor. These organoids can help researchers identify new treatment options and predict how individual patients may respond to standard therapies. By using minimally invasive samples such as blood and urine, patients can participate in research studies more easily, potentially leading to improved outcomes in bladder cancer treatment.
Collapse
Affiliation(s)
- Martina Radić
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Martin Egger
- Department of Urology, Hospital Center Biel, Spitalzentrum Biel, Biel, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland; Department of Urology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Roland Seiler
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland; Department of Urology, Hospital Center Biel, Spitalzentrum Biel, Biel, Switzerland.
| |
Collapse
|
38
|
Yang J, Liu B, Wang Q, Yan H, Li G, Wang X, Shang Z, Ou T, Chen W. Carboxylated mesoporous silica nanoparticle-nucleic acid chimera conjugate-assisted delivery of siRNA and doxorubicin effectively treat drug-resistant bladder cancer. Biomed Pharmacother 2024; 178:117185. [PMID: 39053429 DOI: 10.1016/j.biopha.2024.117185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Chemotherapy is the main treatment for bladder cancer, but drug resistance and side effects limit its application and therapeutic effect. Herein, we constructed doxorubicin (DOX)/COOH-mesoporous silica nanoparticle/polyethylenimine (PEI)/nucleic acid chimeras (DOX/MSN/Chimeras) to reduce the toxicity of chemotherapy drugs and the resistance of bladder cancer cells. Transmission electron microscopy showed that PEI was coated on the DOX/MSN/BSA nanoparticles with a diameter of about 150 nm. DOX/MSN/PEI could control DOX release for over 48 h, and the sudden release rate was significantly lower than DOX/MSN. Immunohistochemical results showed that DOX/MSN/Chimera specifically bound to bladder cancer cells, and markedly inhibited PI3K expression and proliferation of DOX-resistant bladder cancer cells. DOX/MSN/Chimera promoted the apoptosis of drug-resistant bladder cancer cells, which was superior to DOX/MSN/Aptamer or DOX/MSN. We further carried out animal experiments and found that DOX/MSN/Chimera could reduce the volume of transplanted tumors in vivo. Compared with DOX/MSN/Aptamer group, the proliferation rate was significantly decreased and the proportion of apoptotic cells was highly increased. Through the histological observation of kidneys and lungs, we believed that DOX/MSN/Chimera can effectively reduce the damage of chemotherapy drugs to normal tissues. In conclusion, we constructed a COOH-MSN/nucleic acid chimera conjugate for the targeted delivery of siRNA and anti-cancer drugs. Our study provides a new method for personalized and targeted treatment of drug-resistant bladder cancer.
Collapse
Affiliation(s)
- Jintao Yang
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Biao Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Qi Wang
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hao Yan
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Guangping Li
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xu Wang
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhenhua Shang
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Tongwen Ou
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Wen Chen
- Department of Pathology, The 8th Medical Center, Chinese PLA General Hospital, Beijing 100091, China.
| |
Collapse
|
39
|
Zhu X, Trehan R, Xie C. Primary liver cancer organoids and their application to research and therapy. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:195-202. [PMID: 39281720 PMCID: PMC11401492 DOI: 10.1016/j.jncc.2024.06.002] [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: 03/04/2024] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Primary liver cancer is a leading cause of death worldwide. To create advanced treatments for primary liver cancer, studies have utilized models such as 2D cell culture and in vivo animal models. Recent developments in cancer organoids have created the possibility for 3D in vitro cultures that recapitulates the cancer cell structure and operation as well as the tumor microenvironment (TME). However, before organoids can be directly translated to clinical use, tissue processing and culture medium must be standardized with unified protocols to decrease variability in results. Herein, we present the wide variety of published methodologies used to derive liver cancer organoids from patient tumor tissues. Additionally, we summarize validation methodologies for organoids in terms of marker expression levels with immunohistochemistry as well as the presence of mutations and variants through RNA-sequencing. Primary liver cancer organoids have exciting applications allowing for faster drug testing at a larger scale. Primary liver cancer organoids also assisit in uncovering new mechanisms. Through the coculture of different immune cells and cancer organoids, organoids are now better able to recapitulate the liver cancer TME. In addition, it further aids in the investigation of drug development and drug resistance. Lastly, we posit that the usage of liver cancer organoids in animal models provides researchers a methodology to overcome the current limitations of culture systems.
Collapse
Affiliation(s)
- Xiaobin Zhu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Rajiv Trehan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Changqing Xie
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| |
Collapse
|
40
|
Tang X, Berger MF, Solit DB. Precision oncology: current and future platforms for treatment selection. Trends Cancer 2024; 10:781-791. [PMID: 39030146 DOI: 10.1016/j.trecan.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/21/2024]
Abstract
Genomic profiling of hundreds of cancer-associated genes is now a component of routine cancer care. DNA sequencing can identify mutations, mutational signatures, and structural alterations predictive of therapy response and assess for heritable cancer risk, but it has been less useful for identifying predictive biomarkers of sensitivity to cytotoxic chemotherapies, antibody drug conjugates, and immunotherapies. The clinical adoption of molecular profiling platforms such as RNA sequencing better suited to identifying those patients most likely to respond to immunotherapies and drug combinations will be critical to expanding the benefits of precision oncology. This review discusses the potential advantages of innovative molecular and functional profiling platforms designed to replace or complement targeted DNA sequencing and the major hurdles to their clinical adoption.
Collapse
Affiliation(s)
- Xinran Tang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael F Berger
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
41
|
Huang S, Mei Z, Wan A, Zhao M, Qi X. Application and prospect of organoid technology in breast cancer. Front Immunol 2024; 15:1413858. [PMID: 39253075 PMCID: PMC11381393 DOI: 10.3389/fimmu.2024.1413858] [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: 04/08/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer is the most common malignant tumor in women. Due to the high heterogeneity of breast cancer cells, traditional in vitro research models still have major limitations. Therefore, it is urgent to establish an experimental model that can accurately simulate the characteristics of human breast cancer. Breast cancer organoid technology emerged as the times required, that is, to construct tissue analogs with organ characteristics by using a patient's tumor tissue through 3D culture in vitro. Since the breast cancer organoid can fully preserve the histology and genetic characteristics of the original tumor, it provides a reliable model for preclinical drug screening, establishment of breast cancer organoid biobanks, research into the mechanisms of tumor development, and determination of cancer targets. It has promoted personalized treatment for clinical breast cancer patients. This article mainly focuses on recent research progress and applications of organoid technology in breast cancer, discussing the current limitations and prospects of breast cancer organoid technology.
Collapse
Affiliation(s)
- Shanlin Huang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zifan Mei
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Min Zhao
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
42
|
Vollmer P, Amend B, Harland N, Stenzl A, Tsaur I, Maas M, Aicher WK, Walz S. Patient-derived bladder cancer organoids show stable transcript expression along cultivation. World J Urol 2024; 42:468. [PMID: 39110253 DOI: 10.1007/s00345-024-05182-z] [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: 05/20/2024] [Accepted: 07/14/2024] [Indexed: 01/04/2025] Open
Abstract
INTRODUCTION Bladder cancer (BC) is a prevalent malignancy with high recurrence rates. Patient-derived bladder cancer organoids (BCO) pose as a promising approach in both, disease modeling and individualized treatment screening. The aim of this study was to investigate the transcriptomic plasticity in BCOs as a function of cultivation times to define ideal time periods for the applications envisioned. METHODS Tumor samples of three patients with pathologically confirmed non-muscle invasive and muscle-invasive bladder cancer were included in this study and expanded as BCOs. RNA expression was investigated at different time periods of cells in culture using differential gene expression for overall transcript expression and quantitative real-time PCR (qRT-PCR) for pathological relevant markers. RESULTS Differential gene expression of the BCO lines was investigated across passages 1-4, in passages 5-9 and above 9, respectively. Analysis of the entire transcriptome of the respective BCO lines revealed consistent profiles without significant alterations throughout the cultivation and expansion procedure. Notably, key transcripts like TP53, PIK3CA, BRCA1, among others, exhibited stable expression levels in the quantitative RNA analysis during the cultivation period. CONCLUSION The robust transcriptome during BCO cultivation advocates for the use of earlier passages of BCOs in personalized medicine providing a time-efficient drug screening option to accelerate the counseling of patients' treatment options. Higher passages of BCOs still hold the potential in topics demanding for expanded cell masses such as medical device development and others.
Collapse
Affiliation(s)
- Philipp Vollmer
- Center for Medical Research, University of Tuebingen, 72074, Tübingen, Germany
| | - Bastian Amend
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany
| | - Niklas Harland
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany
| | - Igor Tsaur
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany
| | - Moritz Maas
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany
| | - Wilhelm K Aicher
- Center for Medical Research, University of Tuebingen, 72074, Tübingen, Germany
| | - Simon Walz
- Department of Urology, Tuebingen University Hospital, 72076, Tübingen, Germany.
| |
Collapse
|
43
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
44
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
45
|
Yu J, Wang K, Tang Y, Zheng D. Applications and perspectives of tumor organoids in radiobiology (Review). Oncol Rep 2024; 52:100. [PMID: 38904192 PMCID: PMC11223011 DOI: 10.3892/or.2024.8759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
Radiotherapy exhibits significant versatility and efficacy in cancer treatment, thereby playing a crucial role in the field of oncology. However, there remains an urgent need for extensive research on various aspects of radiotherapy, including target selection, damage repair and its combination with immunotherapy. Particularly, the development of in vitro models to replicate in vivo tumor lesion responses is vital. The present study provides a thorough review of the establishment and application of tumor organoids in radiotherapy, aiming to explore their potential impact on cancer treatment.
Collapse
Affiliation(s)
- Jin Yu
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Kailun Wang
- Emergency Department, Panshihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Yongjiang Tang
- Department of Vascular Surgery, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Dalin Zheng
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| |
Collapse
|
46
|
Rahman MM, Wells G, Rantala JK, Helleday T, Muthana M, Danson SJ. In-vitro assays for immuno-oncology drug efficacy assessment and screening for personalized cancer therapy: scopes and challenges. Expert Rev Clin Immunol 2024; 20:821-838. [PMID: 38546609 DOI: 10.1080/1744666x.2024.2336583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Immunotherapies have revolutionized cancer treatment, but often fail to produce desirable therapeutic outcomes in all patients. Due to the inter-patient heterogeneity and complexity of the tumor microenvironment, personalized treatment approaches are gaining demand. Researchers have long been using a range of in-vitro assays including 2D models, organoid co-cultures, and cancer-on-a-chip platforms for cancer drug screening. A comparative analysis of these assays with their suitability, high-throughput capacity, and clinical translatability is required for optimal translational use. AREAS COVERED The review summarized in-vitro platforms with their comparative advantages and limitations including construction strategies, and translational potential for immuno-oncology drug efficacy assessment. We also discussed end-point analysis strategies so that researchers can contextualize their usefulness and optimally design experiments for personalized immunotherapy efficacy prediction. EXPERT OPINION Researchers developed several in-vitro platforms that can provide information on personalized immunotherapy efficacy from different angles. Image-based assays are undoubtedly more suitable to gather a wide range of information including cellular morphology and phenotypical behaviors but need significant improvement to overcome issues including background noise, sample preparation difficulty, and long duration of experiment. More studies and clinical trials are needed to resolve these issues and validate the assays before they can be used in real-life scenarios.
Collapse
Affiliation(s)
- Md Marufur Rahman
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Directorate General of Health Services, Dhaka, Bangladesh
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Juha K Rantala
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Misvik Biology Ltd, Turku, Finland
| | - Thomas Helleday
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Department of Oncology-Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Munitta Muthana
- Nanobug Oncology Sheffield, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Sarah J Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
47
|
Wang Z, Hulikova A, Swietach P. Innovating cancer drug discovery with refined phenotypic screens. Trends Pharmacol Sci 2024; 45:723-738. [PMID: 39013672 DOI: 10.1016/j.tips.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
Before molecular pathways in cancer were known to a depth that could predict targets, drug development relied on phenotypic screening, where the effectiveness of candidate chemicals is judged from functional readouts without considering the mechanisms of action. The unraveling of tumor-specific pathways has brought targets for molecularly driven drug discovery, but precedents in the field have shown that awareness of pathways does not necessarily predict therapeutic efficacy, and many cancers still lack druggable targets. Phenotypic screening therefore retains a niche in drug development where a targeted approach is not informative. We analyze the unique advantages of phenotypic screens, and how technological advances have improved their discovery power. Notable advances include the use of larger biological panels and refined protocols that address the disease-relevance and increase data content with imaging and omic approaches.
Collapse
Affiliation(s)
- Zhenyi Wang
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
48
|
Wu H, Wang W, Zhang Y, Chen Y, Shan C, Li J, Jia Y, Li C, Du C, Cai Y, Zhang Y, Zhang S, Wu F. Establishment of patient-derived organoids for guiding personalized therapies in breast cancer patients. Int J Cancer 2024; 155:324-338. [PMID: 38533706 DOI: 10.1002/ijc.34931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/28/2024]
Abstract
Breast cancer has become the most commonly diagnosed cancer. The intra- and interpatient heterogeneity induced a considerable variation in treatment efficacy. There is an urgent requirement for preclinical models to anticipate the effectiveness of individualized drug responses. Patient-derived organoids (PDOs) can accurately recapitulate the architecture and biological characteristics of the origin tumor, making them a promising model that can overtake many limitations of cell lines and PDXs. However, it is still unclear whether PDOs-based drug testing can benefit breast cancer patients, particularly those with tumor recurrence or treatment resistance. Fresh tumor samples were surgically resected for organoid culture. Primary tumor samples and PDOs were subsequently subjected to H&E staining, immunohistochemical (IHC) analysis, and whole-exome sequencing (WES) to make comparisons. Drug sensitivity tests were performed to evaluate the feasibility of this model for predicting patient drug response in clinical practice. We established 75 patient-derived breast cancer organoid models. The results of H&E staining, IHC, and WES revealed that PDOs inherited the histologic and genetic characteristics of their parental tumor tissues. The PDOs successfully predicted the patient's drug response, and most cases exhibited consistency between PDOs' drug susceptibility test results and the clinical response of the matched patient. We conclude that the breast cancer organoids platform can be a potential preclinical tool used for the selection of effective drugs and guided personalized therapies for patients with advanced breast cancer.
Collapse
Affiliation(s)
- Huizi Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Weiwei Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yinbin Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yinxi Chen
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Changyou Shan
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jia Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yiwei Jia
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chaofan Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chong Du
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yifan Cai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yu Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Fei Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
49
|
Thorel L, Perréard M, Florent R, Divoux J, Coffy S, Vincent A, Gaggioli C, Guasch G, Gidrol X, Weiswald LB, Poulain L. Patient-derived tumor organoids: a new avenue for preclinical research and precision medicine in oncology. Exp Mol Med 2024; 56:1531-1551. [PMID: 38945959 PMCID: PMC11297165 DOI: 10.1038/s12276-024-01272-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/18/2024] [Accepted: 04/14/2024] [Indexed: 07/02/2024] Open
Abstract
Over the past decade, the emergence of patient-derived tumor organoids (PDTOs) has broadened the repertoire of preclinical models and progressively revolutionized three-dimensional cell culture in oncology. PDTO can be grown from patient tumor samples with high efficiency and faithfully recapitulates the histological and molecular characteristics of the original tumor. Therefore, PDTOs can serve as invaluable tools in oncology research, and their translation to clinical practice is exciting for the future of precision medicine in oncology. In this review, we provide an overview of methods for establishing PDTOs and their various applications in cancer research, starting with basic research and ending with the identification of new targets and preclinical validation of new anticancer compounds and precision medicine. Finally, we highlight the challenges associated with the clinical implementation of PDTO, such as its representativeness, success rate, assay speed, and lack of a tumor microenvironment. Technological developments and autologous cocultures of PDTOs and stromal cells are currently ongoing to meet these challenges and optimally exploit the full potential of these models. The use of PDTOs as standard tools in clinical oncology could lead to a new era of precision oncology in the coming decade.
Collapse
Grants
- AP-RM-19-020 Fondation de l'Avenir pour la Recherche Médicale Appliquée (Fondation de l'Avenir)
- PJA20191209649 Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Ligue Contre le Cancer
- ORGAPRED Ligue Contre le Cancer
- 3D-Hub Canceropôle PACA (Canceropole PACA)
- Pré-néo 2019-188 Institut National Du Cancer (French National Cancer Institute)
- Conseil Régional de Haute Normandie (Upper Normandy Regional Council)
- GIS IBiSA, Cancéropôle Nord-Ouest (ORGRAFT project), the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (ORGAVADS project), the Fonds de dotation Patrick de Brou de Laurière (ORGAVADS project),and Normandy County Council (ORGATHEREX project).
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), Etat-région
- GIS IBiSA, Region Sud
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), and Normandy County Council (ORGAPRED, PLATONUS ONE, POLARIS, and EQUIP’INNOV projects).
Collapse
Affiliation(s)
- Lucie Thorel
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Marion Perréard
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Department of Head and Neck Surgery, Caen University Hospital, Caen, France
| | - Romane Florent
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Jordane Divoux
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Sophia Coffy
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Audrey Vincent
- CNRS UMR9020, INSERM U1277, CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, CNRS, Inserm, CHU Lille, Lille, France
| | - Cédric Gaggioli
- CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, CNRS University Côte d'Azur, Nice, France
| | - Géraldine Guasch
- CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Epithelial Stem Cells and Cancer Team, Aix-Marseille University, Marseille, France
| | - Xavier Gidrol
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Louis-Bastien Weiswald
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| | - Laurent Poulain
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| |
Collapse
|
50
|
Mazzucchelli S, Signati L, Messa L, Franceschini A, Bonizzi A, Castagnoli L, Gasparini P, Consolandi C, Mangano E, Pelucchi P, Cifola I, Camboni T, Severgnini M, Villani L, Tagliaferri B, Carelli S, Pupa SM, Cereda C, Corsi F. Breast cancer patient-derived organoids for the investigation of patient-specific tumour evolution. Cancer Cell Int 2024; 24:220. [PMID: 38926706 PMCID: PMC11210105 DOI: 10.1186/s12935-024-03375-5] [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: 12/14/2023] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND A reliable preclinical model of patient-derived organoids (PDOs) was developed in a case study of a 69-year-old woman diagnosed with breast cancer (BC) to investigate the tumour evolution before and after neoadjuvant chemotherapy and surgery. The results were achieved due to the development of PDOs from tissues collected before (O-PRE) and after (O-POST) treatment. METHODS PDO cultures were characterized by histology, immunohistochemistry (IHC), transmission electron microscopy (TEM), scanning electron microscopy (SEM), confocal microscopy, flow cytometry, real-time PCR, bulk RNA-seq, single-cell RNA sequencing (scRNA-seq) and drug screening. RESULTS Both PDO cultures recapitulated the histological and molecular profiles of the original tissues, and they showed typical mammary gland organization, confirming their reliability as a personalized in vitro model. Compared with O-PRE, O-POST had a greater proliferation rate with a significant increase in the Ki67 proliferation index. Moreover O-POST exhibited a more stem-like and aggressive phenotype, with increases in the CD24low/CD44low and EPCAMlow/CD49fhigh cell populations characterized by increased tumour initiation potential and multipotency and metastatic potential in invasive lobular carcinoma. Analysis of ErbB receptor expression indicated a decrease in HER-2 expression coupled with an increase in EGFR expression in O-POST. In this context, deregulation of the PI3K/Akt signalling pathway was assessed by transcriptomic analysis, confirming the altered transcriptional profile. Finally, transcriptomic single-cell analysis identified 11 cell type clusters, highlighting the selection of the luminal component and the decrease in the number of Epithelial-mesenchymal transition cell types in O-POST. CONCLUSION Neoadjuvant treatment contributed to the enrichment of cell populations with luminal phenotypes that were more resistant to chemotherapy in O-POST. PDOs represent an excellent 3D cell model for assessing disease evolution.
Collapse
Affiliation(s)
- Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy.
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy
| | - Letizia Messa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133, Milan, Italy
- Pediatric Research Center "Romeo and Enrica Invernizzi", Università di Milano, 20157, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Alma Franceschini
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Arianna Bonizzi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - Lorenzo Castagnoli
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Patrizia Gasparini
- Epigenomics and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Clarissa Consolandi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Eleonora Mangano
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Tania Camboni
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Marco Severgnini
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Laura Villani
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | | | - Stephana Carelli
- Pediatric Research Center "Romeo and Enrica Invernizzi", Università di Milano, 20157, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Serenella M Pupa
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy.
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy.
| |
Collapse
|