1
|
Garimella A, Ghosh SB, Bandyopadhyay-Ghosh S. Biomaterials for bone tissue engineering: achievements to date and future directions. Biomed Mater 2024; 20:012001. [PMID: 39577395 DOI: 10.1088/1748-605x/ad967c] [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: 06/26/2024] [Accepted: 11/22/2024] [Indexed: 11/24/2024]
Abstract
Advancement in medicine and technology has resulted into prevention of countless deaths and increased life span. However, it is important to note that, the modern lifestyle has altered the food habits, witnessed increased life-style stresses and road accidents leading to several health complications and one of the primary victims is the bone health. More often than ever, healthcare professionals encounter cases of massive bone fracture, bone loss and generation of critical sized bone defects. Surgical interventions, through the use of bone grafting techniques are necessary in such cases. Natural bone grafts (allografts, autografts and xenografts) however, have major drawbacks in terms of delayed rehabilitation, lack of appropriate donors, infection and morbidity that shifted the focus of several investigators to the direction of synthetic bone grafts. By employing biomaterials that are based on bone tissue engineering (BTE), synthetic bone grafts provide a more biologically acceptable approach to establishing the phases of bone healing. In BTE, various materials are utilized to support and enhance bone regeneration. Biodegradable polymers like poly-(lactic acid), poly-(glycolic acid), and poly-(ϵ-caprolactone) are commonly used for their customizable mechanical properties and ability to degrade over time, allowing for natural bone growth. PEG is employed in hydrogels to promote cell adhesion and growth. Ceramics, such as hydroxyapatite and beta-tricalcium phosphate (β-TCP) mimic natural bone mineral and support bone cell attachment, withβ-TCP gradually resorbing as new bone forms. Composite materials, including polymer-ceramic and polymer-glasses, combine the benefits of both polymers and ceramics/glasses to offer enhanced mechanical and biological properties. Natural biomaterials like collagen, gelatin, and chitosan provide a natural matrix for cell attachment and tissue formation, with chitosan also offering antimicrobial properties. Hybrid materials such as decellularized bone matrix retain natural bone structure and biological factors, while functionalized scaffolds incorporate growth factors or bioactive molecules to further stimulate bone healing and integration. The current review article provides the critical insights on several biomaterials that could yield to revolutionary improvements in orthopedic medical fields. The introduction section of this article focuses on the statistical information on the requirements of various bone scaffolds globally and its impact on economy. In the later section, anatomy of the human bone, defects and diseases pertaining to human bone, and limitations of natural bone scaffolds and synthetic bone scaffolds were detailed. Biopolymers, bioceramics, and biometals-based biomaterials were discussed in further depth in the sections that followed. The article then concludes with a summary addressing the current trends and the future prospects of potential bone transplants.
Collapse
Affiliation(s)
- Adithya Garimella
- Department of Mechanical and Industrial Engineering, Manipal Institute of Technology Bengaluru, Manipal Academy of Higher Education, Manipal, India
| | - Subrata Bandhu Ghosh
- Engineered Biomedical Materials Research and Innovation Centre (EnBioMatRIC), Department of Mechanical Engineering, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Sanchita Bandyopadhyay-Ghosh
- Engineered Biomedical Materials Research and Innovation Centre (EnBioMatRIC), Department of Mechanical Engineering, Manipal University Jaipur, Jaipur, Rajasthan, India
| |
Collapse
|
2
|
Kojima Y, Yoshida H, Okuya T, Okuma HS, Nishikawa T, Tanioka M, Sudo K, Noguchi E, Shimoi T, Tamura K, Tanase Y, Uno M, Ishikawa M, Arakaki M, Ichikawa H, Yagishita S, Hamada A, Fujiwara Y, Yonemori K, Kato T. Therapeutic target biomarkers of patient-derived xenograft models of gastric-type cervical adenocarcinoma. Gynecol Oncol Rep 2023; 50:101302. [PMID: 38054200 PMCID: PMC10694048 DOI: 10.1016/j.gore.2023.101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/04/2023] [Accepted: 11/04/2023] [Indexed: 12/07/2023] Open
Abstract
Background Most cervical adenocarcinomas are associated with human papillomavirus (HPV). Gastric-type cervical adenocarcinoma (GAS), an HPV-independent adenocarcinoma, shows an aggressive clinical feature, resulting in a poor prognosis. Resistance to chemotherapy poses a difficulty in managing patients with metastatic GAS. We aimed to establish patient-derived xenografts (PDXs) of tumors from two patients with GAS and evaluated protein biomarkers for drug development using immunohistochemistry. Methods Two PDXs were established 78 and 48 days after transplanting the patient's tumor tissues into immunodeficient mice, respectively. PDX and patient's tumor samples were stained for HER2, HER3, PMS2, MSH6, PanTrk, and ARID1A to evaluate biomarkers for therapeutic targets. In addition, whole exome sequencing and RNA sequencing were performed on available samples. Results The pathological findings in morphological features and immunohistochemical profiles from the established PDXs were similar to those from the patients' surgical tumor specimens. HER3 was overexpressed in the patient's tumors, and the corresponding PDX tumors and HER2 was weakly stained in both types of tumor samples. In all PDX and patient tumor samples, PMS2, MSH6, and ARID1A were retained, and PanTrk was not expressed. In addition, a total of 10 samples, including tumor tissue samples from 8 other GAS patients, were evaluated for HER3 expression scores, all of which were 2 + or higher. Conclusions In summary, we evaluated biomarkers for therapeutic targets using newly established PDX models of GAS. Frequent HER3 overexpression and HER2 expression in GAS tumors suggest the possibility of new treatments for patients with GAS by targeting HER3 and HER2.
Collapse
Affiliation(s)
- Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
- Department of Molecular Pharmacology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Toshihiro Okuya
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Hitomi S Okuma
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Maki Tanioka
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Emi Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Kenji Tamura
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Yasuhito Tanase
- Department of Gynecology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Masaya Uno
- Department of Gynecology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Mitsuya Ishikawa
- Department of Gynecology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Motoko Arakaki
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
- Department of Clinical Genomics, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Hitoshi Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Shigehiro Yagishita
- Department of Molecular Pharmacology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Akinobu Hamada
- Department of Molecular Pharmacology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
3
|
Zeng M, Ruan Z, Tang J, Liu M, Hu C, Fan P, Dai X. Generation, evolution, interfering factors, applications, and challenges of patient-derived xenograft models in immunodeficient mice. Cancer Cell Int 2023; 23:120. [PMID: 37344821 DOI: 10.1186/s12935-023-02953-3] [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: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 06/23/2023] Open
Abstract
Establishing appropriate preclinical models is essential for cancer research. Evidence suggests that cancer is a highly heterogeneous disease. This follows the growing use of cancer models in cancer research to avoid these differences between xenograft tumor models and patient tumors. In recent years, a patient-derived xenograft (PDX) tumor model has been actively generated and applied, which preserves both cell-cell interactions and the microenvironment of tumors by directly transplanting cancer tissue from tumors into immunodeficient mice. In addition to this, the advent of alternative hosts, such as zebrafish hosts, or in vitro models (organoids and microfluidics), has also facilitated the advancement of cancer research. However, they still have a long way to go before they become reliable models. The development of immunodeficient mice has enabled PDX to become more mature and radiate new vitality. As one of the most reliable and standard preclinical models, the PDX model in immunodeficient mice (PDX-IM) exerts important effects in drug screening, biomarker development, personalized medicine, co-clinical trials, and immunotherapy. Here, we focus on the development procedures and application of PDX-IM in detail, summarize the implications that the evolution of immunodeficient mice has brought to PDX-IM, and cover the key issues in developing PDX-IM in preclinical studies.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zijing Ruan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaxi Tang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Maozhu Liu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Fan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xinhua Dai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Structural and Functional Thymic Biomarkers Are Involved in the Pathogenesis of Thymic Epithelial Tumors: An Overview. IMMUNO 2022. [DOI: 10.3390/immuno2020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The normal human thymus originates from the third branchial cleft as two paired anlages that descend into the thorax and fuse on the midline of the anterior–superior mediastinum. Alongside the epithelial and lymphoid components, different types of lymphoid accessory cells, stromal mesenchymal and endothelial cells migrate to, or develop in, the thymus. After reaching maximum development during early postnatal life, the human thymus decreases in size and lymphocyte output drops with age. However, thymic immunological functions persist, although they deteriorate progressively. Several major techniques were fundamental to increasing the knowledge of thymic development and function during embryogenesis, postnatal and adult life; these include immunohistochemistry, immunofluorescence, flow cytometry, in vitro colony assays, transplantation in mice models, fetal organ cultures (FTOC), re-aggregated thymic organ cultures (RTOC), and whole-organ thymic scaffolds. The thymic morphological and functional characterization, first performed in the mouse, was then extended to humans. The purpose of this overview is to provide a report on selected structural and functional biomarkers of thymic epithelial cells (TEC) involved in thymus development and lymphoid cell maturation, and on the historical aspects of their characterization, with particular attention being paid to biomarkers also involved in Thymic Epithelial Tumor (TET) pathogenesis. Moreover, a short overview of targeted therapies in TET, based on currently available experimental and clinical data and on potential future advances will be proposed.
Collapse
|
5
|
Chen J, Jin Y, Li S, Qiao C, Peng X, Li Y, Gu Y, Wang W, You Y, Yin J, Shan Y, Wang YX, Qin M, Li H, Cai Y, Dong Y, Peng S, Pan L. Patient-Derived Xenografts Are a Reliable Preclinical Model for the Personalized Treatment of Epithelial Ovarian Cancer. Front Oncol 2021; 11:744256. [PMID: 34671560 PMCID: PMC8522495 DOI: 10.3389/fonc.2021.744256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
To generate robust patient-derived xenograft (PDX) models for epithelial ovarian cancer (EOC), analyze the resemblance of PDX models to the original tumors, and explore factors affecting engraftment rates, fresh cancer tissues from a consecutive cohort of 158 patients with EOC were collected to construct subcutaneous PDX models. Paired samples of original tumors and PDX tumors were compared at the genome, transcriptome, protein levels, and the platinum-based chemotherapy response was evaluated to ensure the reliability of the PDXs. Univariate and multivariate analyses were used to determine the factors affecting the engraftment rates. The engraftment success rate was 58.23% (92/158) over 3–6 months. The Ki-67 index and receiving neoadjuvant chemotherapy can affect the engraftment rate in primary patients. The PDX models generated in this study were found to retain the histomorphology, protein expression, and genetic alteration patterns of the original tumors, despite the transcriptomic differences observed. Clinically, the PDX models demonstrated a high degree of similarity with patients in terms of the chemotherapy response and could predict prognosis. Thus, the PDX model can be considered a promising and reliable preclinical tool for personalized and precise treatment.
Collapse
Affiliation(s)
- Jiayu Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Ying Jin
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Siyi Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Cui Qiao
- National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China.,Beijing IDMO Co., Ltd., Beijing, China
| | - Xinxin Peng
- The Bioinformatics Department, Precision Scientific (Beijing) Co., Ltd., Beijing, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Yu Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Yan You
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Jie Yin
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Ying Shan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Yong-Xue Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Meng Qin
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Hongyue Li
- The Bioinformatics Department, Precision Scientific (Beijing) Co., Ltd., Beijing, China
| | - Yan Cai
- Department of Obstetrics and Gynecology, Beijing Cancer Hospital, Beijing, China
| | - Yu Dong
- The Medical Department, Precision Scientific (Beijing) Co., Ltd., Beijing, China
| | | | - Lingya Pan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| |
Collapse
|
6
|
Stiffness increases with myofibroblast content and collagen density in mesenchymal high grade serous ovarian cancer. Sci Rep 2021; 11:4219. [PMID: 33603134 PMCID: PMC7892556 DOI: 10.1038/s41598-021-83685-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/05/2021] [Indexed: 01/31/2023] Open
Abstract
Women diagnosed with high-grade serous ovarian cancers (HGSOC) are still likely to exhibit a bad prognosis, particularly when suffering from HGSOC of the Mesenchymal molecular subtype (50% cases). These tumors show a desmoplastic reaction with accumulation of extracellular matrix proteins and high content of cancer-associated fibroblasts. Using patient-derived xenograft mouse models of Mesenchymal and Non-Mesenchymal HGSOC, we show here that HGSOC exhibit distinct stiffness depending on their molecular subtype. Indeed, tumor stiffness strongly correlates with tumor growth in Mesenchymal HGSOC, while Non-Mesenchymal tumors remain soft. Moreover, we observe that tumor stiffening is associated with high stromal content, collagen network remodeling, and MAPK/MEK pathway activation. Furthermore, tumor stiffness accompanies a glycolytic metabolic switch in the epithelial compartment, as expected based on Warburg's effect, but also in stromal cells. This effect is restricted to the central part of stiff Mesenchymal tumors. Indeed, stiff Mesenchymal tumors remain softer at the periphery than at the core, with stromal cells secreting high levels of collagens and showing an OXPHOS metabolism. Thus, our study suggests that tumor stiffness could be at the crossroad of three major processes, i.e. matrix remodeling, MEK activation and stromal metabolic switch that might explain at least in part Mesenchymal HGSOC aggressiveness.
Collapse
|
7
|
Graim K, Gorenshteyn D, Robinson DG, Carriero NJ, Cahill JA, Chakrabarti R, Goldschmidt MH, Durham AC, Funk J, Storey JD, Kristensen VN, Theesfeld CL, Sorenmo KU, Troyanskaya OG. Modeling molecular development of breast cancer in canine mammary tumors. Genome Res 2021; 31:337-347. [PMID: 33361113 PMCID: PMC7849403 DOI: 10.1101/gr.256388.119] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/17/2020] [Indexed: 12/30/2022]
Abstract
Understanding the changes in diverse molecular pathways underlying the development of breast tumors is critical for improving diagnosis, treatment, and drug development. Here, we used RNA-profiling of canine mammary tumors (CMTs) coupled with a robust analysis framework to model molecular changes in human breast cancer. Our study leveraged a key advantage of the canine model, the frequent presence of multiple naturally occurring tumors at diagnosis, thus providing samples spanning normal tissue and benign and malignant tumors from each patient. We showed human breast cancer signals, at both expression and mutation level, are evident in CMTs. Profiling multiple tumors per patient enabled by the CMT model allowed us to resolve statistically robust transcription patterns and biological pathways specific to malignant tumors versus those arising in benign tumors or shared with normal tissues. We showed that multiple histological samples per patient is necessary to effectively capture these progression-related signatures, and that carcinoma-specific signatures are predictive of survival for human breast cancer patients. To catalyze and support similar analyses and use of the CMT model by other biomedical researchers, we provide FREYA, a robust data processing pipeline and statistical analyses framework.
Collapse
Affiliation(s)
- Kiley Graim
- Flatiron Institute, Simons Foundation, New York, New York 10010, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
| | - Dmitriy Gorenshteyn
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - David G Robinson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, New Jersey 08544, USA
| | | | - James A Cahill
- Laboratory of the Neurogenetics of Language, Rockefeller University, New York, New York 10065, USA
| | - Rumela Chakrabarti
- Department of Biomedical Sciences and the Penn Vet Cancer Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael H Goldschmidt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Amy C Durham
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Julien Funk
- Flatiron Institute, Simons Foundation, New York, New York 10010, USA
| | - John D Storey
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, New Jersey 08544, USA
| | - Vessela N Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0310 Oslo, Norway
| | - Chandra L Theesfeld
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
| | - Karin U Sorenmo
- Department of Biomedical Sciences and the Penn Vet Cancer Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Olga G Troyanskaya
- Flatiron Institute, Simons Foundation, New York, New York 10010, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Department of Computer Science, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
8
|
Yoshida K, Yokoi A, Sugiyama M, Oda S, Kitami K, Tamauchi S, Ikeda Y, Yoshikawa N, Nishino K, Niimi K, Suzuki S, Kikkawa F, Yokoi T, Kajiyama H. Expression of the chrXq27.3 miRNA cluster in recurrent ovarian clear cell carcinoma and its impact on cisplatin resistance. Oncogene 2021; 40:1255-1268. [PMID: 33420363 PMCID: PMC7892337 DOI: 10.1038/s41388-020-01595-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Ovarian clear cell carcinoma (OCCC) is a histological subtype of epithelial ovarian cancer and exhibits dismal prognosis due to chemoresistance. Moreover, only few effective therapeutic options exist for patients with recurrent OCCC, and an understanding of its molecular characteristics is essential for the development of novel therapeutic approaches. In the present study, we investigated unique MicroRNAs (miRNA) profiles in recurrent/metastatic OCCC and the role of miRNAs in cisplatin resistance. Comprehensive miRNA sequencing revealed that expression of several miRNAs, including miR-508-3p, miR-509-3p, miR-509-3-5p, and miR-514a-3p was remarkably less in recurrent cancer tissues when compared with that in paired primary cancer tissues. These miRNAs are located in the chrXq27.3 region on the genome. Moreover, its expression was negative in omental metastases in two patients with advanced OCCC. In vitro analyses revealed that overexpression of miR-509-3p and miR-509-3-5p reversed cisplatin resistance and yes-associated protein 1 (YAP1) was partially responsible for the resistance. Immunohistochemistry revealed that YAP1 expression was inversely correlated with the chrXq27.3 miRNA cluster expression. In conclusion, these findings suggest that alteration of the chrXq27.3 miRNA cluster could play a critical role in chemoresistance and miRNAs in the cluster and their target genes can be potential therapeutic targets.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Advanced Research, Nagoya University, Nagoya, Japan.,Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan. .,Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimihiro Nishino
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaoru Niimi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Suzuki
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Song F, Hu B, Cheng JW, Sun YF, Zhou KQ, Wang PX, Guo W, Zhou J, Fan J, Chen Z, Yang XR. Anlotinib suppresses tumor progression via blocking the VEGFR2/PI3K/AKT cascade in intrahepatic cholangiocarcinoma. Cell Death Dis 2020; 11:573. [PMID: 32709873 PMCID: PMC7381674 DOI: 10.1038/s41419-020-02749-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a malignant tumor derived from bile duct epithelium. Its characteristics include an insidious onset and frequent recurrence or metastasis after surgery. Current chemotherapies and molecular target therapies provide only modest survival benefits to patients with ICC. Anlotinib is a novel multi-target tyrosine kinase inhibitor that has good antitumor effects in a variety of solid tumors. However, there are few studies of anlotinib-associated mechanisms and use as a treatment in ICC. In this study using in vitro experiments, we found that anlotinib had significant effects on proliferation inhibition, migration and invasion restraint, and cell-cycle arrestment. Anlotinib treatment affected induction of apoptosis and the mesenchymal-epithelial transition. Patient-derived xenograft models generated directly from patients with ICC revealed that anlotinib treatment dramatically hindered in vivo tumor growth. We also examined anlotinib's mechanism of action using transcriptional profiling. We found that anlotinib treatment might mainly inhibit tumor cell proliferation and invasion and promote apoptosis via cell-cycle arrestment by inactivating the VEGF/PI3K/AKT signaling pathway, as evidenced by significantly decreased phosphorylation levels of these kinases. The activation of vascular endothelial growth factor receptor 2 (VEGFR2) can subsequently activate PI3K/AKT signaling. We identified VEGRF2 as the main target of anlotinib. High VEGFR2 expression might serve as a promising indicator when used to predict a favorable therapeutic response. Taken together, these results indicated that anlotinib had excellent antitumor activity in ICC, mainly via inhibiting the phosphorylation level of VEGFR2 and subsequent inactivation of PIK3/AKT signaling. This work provides evidence and a rationale for using anlotinib to treat patients with ICC in the future.
Collapse
Grants
- National Key Research and Development Program (2016YFF0101405), the National Natural Science Foundation of China (81672839), the National Natural Science Foundation of China (81872355), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA12020103)。
- the Beijing Medical and Health Foundation (YWJKJJHKYJJ-F2093E), the Projects from the Shanghai Science and Technology Commission (19441905000)。
- the National Natural Science Foundation of China (81602543)。
- National Key Research and Development Program (2016YFC0902400), the State Key Program of National Natural Science of China (81830102), the National Natural Science Foundation of China (81572823), the National Natural Science Foundation of China (81772578)。
- the State Key Program of National Natural Science of China (81530077, the National Natural Science Foundation of China (81772551), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA12020105), the Shanghai Municipal Health Commission Collaborative Innovation Cluster Project (2019CXJQ02)。
- the National Natural Science Foundation of China (81871927), the Jiangsu Science and Technology Department Project (BL2014060), the Nantong Hepatobiliary and Pancreatic Surgery Disease Research Center Construction Project (HS2015001)。
Collapse
Affiliation(s)
- Fei Song
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Bo Hu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Jian-Wen Cheng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Yun-Fan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Kai-Qian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Peng-Xiang Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, PR China.
| | - Xin-Rong Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| |
Collapse
|
10
|
Jiang W, Xie S, Liu Y, Zou S, Zhu X. The Application of Patient-Derived Xenograft Models in Gynecologic Cancers. J Cancer 2020; 11:5478-5489. [PMID: 32742495 PMCID: PMC7391187 DOI: 10.7150/jca.46145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, due to the limitations of cell line models and animal models in the preclinical research with insufficient reflecting the physiological situation of humans, patient-derived xenograft (PDX) models of many cancers have been widely developed because of their better representation of the tumor heterogeneity and tumor microenvironment with retention of the cellular complexity, cytogenetics, and stromal architecture. PDX models now have been identified as a powerful tool for determining cancer characteristics, developing new treatment, and predicting drug efficacy. An increase in attempts to generate PDX models in gynecologic cancers has emerged in recent years to understand tumorigenesis. Hence, this review summarized the generation of PDX models and engraftment success of PDX models in gynecologic cancers. Furthermore, we illustrated the similarity between PDX model and original tumor, and described preclinical utilization of PDX models in gynecologic cancers. It would help supply better personalized therapy for gynecologic cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| |
Collapse
|
11
|
Yoshida K, Yokoi A, Kagawa T, Oda S, Hattori S, Tamauchi S, Ikeda Y, Yoshikawa N, Nishino K, Utsumi F, Niimi K, Suzuki S, Shibata K, Kajiyama H, Yokoi T, Kikkawa F. Unique miRNA profiling of squamous cell carcinoma arising from ovarian mature teratoma: comprehensive miRNA sequence analysis of its molecular background. Carcinogenesis 2020; 40:1435-1444. [PMID: 31353396 DOI: 10.1093/carcin/bgz135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/23/2019] [Accepted: 07/27/2019] [Indexed: 12/18/2022] Open
Abstract
Owing to its rarity, the carcinogenesis and molecular biological characteristics of squamous cell carcinoma arising from mature teratoma remain unclear. This study aims to elucidate the molecular background of malignant transformation from the aspects of microRNA (miRNA) profiling. We examined 7 patients with squamous cell carcinoma and 20 patients with mature teratoma and extracted their total RNA from formalin-fixed paraffin-embedded tissues. Then we prepared small RNA libraries and performed comprehensive miRNA sequencing. Heatmap and principal component analysis revealed markedly different miRNA profiling in cancer, normal ovarian and mature teratoma tissues. Then we narrowed down cancer-related miRNAs, comparing paired-cancer and normal ovaries. Comparisons of cancer and mature teratoma identified two markedly upregulated miRNAs (miR-151a-3p and miR-378a-3p) and two markedly downregulated miRNAs (miR-26a-5p and miR-99a-5p). In addition, these findings were validated in fresh cancer tissues of patient-derived xenograft (PDX) models. Moreover, several miRNAs, including miR-151a-3p and miR-378a-3p, were elevated in the murine plasma when tumor tissues were enlarged although miR-26a-5p and miR-99a-5p were not elucidated in the murine plasma. Finally, we performed target prediction and functional annotation analysis in silico and indicated that targets genes of these miRNAs markedly correlated with cancer-related pathways, including 'pathway in cancer' and 'cell cycle'. In conclusion, this is the first study on miRNA sequencing for squamous cell carcinoma arising from mature teratoma. The study identified four cancer-related miRNAs that were considered to be related to the feature of malignant transformation. Moreover, miRNAs circulating in the murine plasma of the PDX model could be novel diagnostic biomarkers.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Takumi Kagawa
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satomi Hattori
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimihiro Nishino
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumi Utsumi
- Department of Obstetrics and Gynecology, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Kaoru Niimi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Suzuki
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyosumi Shibata
- Department of Obstetrics and Gynecology, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
12
|
Lin G, Zhao R, Wang Y, Han J, Gu Y, Pan Y, Ren C, Ren S, Xu C. Dynamic analysis of N-glycomic and transcriptomic changes in the development of ovarian cancer cell line A2780 to its three cisplatin-resistant variants. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:289. [PMID: 32355733 PMCID: PMC7186709 DOI: 10.21037/atm.2020.03.12] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Platinum resistance development is a dynamic process that occurs during continuous chemotherapy and contributes to high mortality in ovarian cancer. Abnormal glycosylation has been reported in platinum resistance. Many studies on platinum resistance have been performed, but few of them have investigated platinum resistance-associated glycans based on N-glycomics. Moreover, glycomic alterations during platinum resistance development in ovarian cancer are rarely reported. Therefore, the objective of this study was to determine platinum resistance-related N-glycans in ovarian cancer cells during continuous exposure to cisplatin. These glycans might be involved in the mechanism of platinum resistance and serve as biomarkers to monitor its development. Methods This study mimicked the development of platinum resistance in ovarian cancer by continuously exposing A2780 cells to cisplatin. Cisplatin-resistant variants were confirmed by higher half maximal inhibitory concentration (IC50) values and increased P-glycoprotein (ABCB1, P-gp) expression compared to A2780 cells. Analysis of dynamic N-glycomic changes during the development of platinum resistance in cisplatin-resistant variants was performed with MALDI-time-of-flight (TOF)-MS combined with ethyl esterification derivatization, which were used to discriminate between α2,3- and α2,6-linkage N-acetylneuraminic acid. N-glycan alterations were further validated on a glycotransferase level via transcriptome sequencing and real-time PCR (RT-PCR). Results Compared to the A2780 cells, MS analysis indicated that α2,3-linked sialic structures and N-glycan gal-ratios were significantly higher, while fucosylated glycans were lower in three cisplatin-resistant variants. Transcriptome sequencing and RT-PCR showed that gene expression of ST3GAL6 and MGAT4A increased, while gene expression of FUT11, FUT1, GMDS, and B4GALT5 decreased in three cisplatin-resistant variants. Conclusions Analysis of N-glycans and glycogene expression showed that α2,3-linked sialic structures might serve as biomarkers to monitor the development of platinum resistance and to guide individualized treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Guiling Lin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Ran Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Yisheng Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Jing Han
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yong Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiqing Pan
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Changhao Ren
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shifang Ren
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Congjian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200032, China.,Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
13
|
Development of a near infrared protein nanoprobe targeting Thomsen-Friedenreich antigen for intraoperative detection of submillimeter nodules in an ovarian peritoneal carcinomatosis mouse model. Biomaterials 2020; 241:119908. [PMID: 32126396 DOI: 10.1016/j.biomaterials.2020.119908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
The epithelial ovarian cancer is one of the most lethal gynecological malignancy due to its late diagnostic and many relapses observed after first line of treatment. Once diagnose, the most important prognostic factor is the completeness of cytoreductive surgery. To achieve this goal, surgeons have to pinpoint and remove nodules, especially the smallest nodules. Recent advances in fluorescence-guided surgery led us to develop a recombinant lectin as a nanoprobe for the microscopic detection of nodules in the peritoneal cavity of tumor-bearing mice. This lectin has an intrinsic specificity for a carcinoma-associated glycan biomarker, the Thomsen-Friedenreich antigen. In this study, after its labelling by a near infrared dye, we first demonstrated that this nanoprobe allowed indirect detection of nodules already implanted in the peritoneal cavity, through tumor microenvironment targeting. Secondly, in a protocol mimicking the scattering of cells during surgery, we obtained a direct and long-lasting detection of tumor cells in vivo. This lectin as already been described as a nanocontainer able to do targeted delivery of a therapeutic compound to carcinoma cells. Future developments will focus on the combination of the nanoprobe and nanocontainer aspects in an intraperitoneal nanotheranostic approach.
Collapse
|
14
|
Espedal H, Fonnes T, Fasmer KE, Krakstad C, Haldorsen IS. Imaging of Preclinical Endometrial Cancer Models for Monitoring Tumor Progression and Response to Targeted Therapy. Cancers (Basel) 2019; 11:cancers11121885. [PMID: 31783595 PMCID: PMC6966645 DOI: 10.3390/cancers11121885] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Endometrial cancer is the most common gynecologic malignancy in industrialized countries. Most patients are cured by surgery; however, about 15% of the patients develop recurrence with limited treatment options. Patient-derived tumor xenograft (PDX) mouse models represent useful tools for preclinical evaluation of new therapies and biomarker identification. Preclinical imaging by magnetic resonance imaging (MRI), positron emission tomography-computed tomography (PET-CT), single-photon emission computed tomography (SPECT) and optical imaging during disease progression enables visualization and quantification of functional tumor characteristics, which may serve as imaging biomarkers guiding targeted therapies. A critical question, however, is whether the in vivo model systems mimic the disease setting in patients to such an extent that the imaging biomarkers may be translatable to the clinic. The primary objective of this review is to give an overview of current and novel preclinical imaging methods relevant for endometrial cancer animal models. Furthermore, we highlight how these advanced imaging methods depict pathogenic mechanisms important for tumor progression that represent potential targets for treatment in endometrial cancer.
Collapse
Affiliation(s)
- Heidi Espedal
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (H.E.); (I.S.H.)
| | - Tina Fonnes
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (T.F.); (C.K.)
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Kristine E. Fasmer
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (T.F.); (C.K.)
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ingfrid S. Haldorsen
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (H.E.); (I.S.H.)
| |
Collapse
|
15
|
Taibi A, Albouys J, Jacques J, Perrin ML, Yardin C, Durand Fontanier S, Bardet SM. Comparison of implantation sites for the development of peritoneal metastasis in a colorectal cancer mouse model using non-invasive bioluminescence imaging. PLoS One 2019; 14:e0220360. [PMID: 31365553 PMCID: PMC6668798 DOI: 10.1371/journal.pone.0220360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/15/2019] [Indexed: 12/18/2022] Open
Abstract
The development of cancer mouse models is still needed for the identification and preclinical validation of novel therapeutic targets in colorectal cancer, which is the third leading cause of cancer-related deaths in Europe. The purpose of this study was to determine the most accurate tumour cell injection method to obtain suitable peritoneal metastasis (PM) for subsequent therapeutic treatments. Here, we grafted murine colon carcinoma CT-26 cells expressing luciferase into immunocompetent BALB-c mice by intravenous injection (IV group), subcutaneous injection (SC group), intraperitoneal injection after peritoneal scratching (A group) or intraperitoneal injection alone (IP group). Tumour growth was monitored by bioluminescence during the first 15 days post-grafting. The peritoneal carcinomatosis index was evaluated macroscopically, histology, immunohistochemistry and multiphoton microscopy were performed in peritoneal tumour tissue. Upon implantation, no tumour growth was observed in the IV group, similar to the non-injected group. Both the IP and SC groups showed intermediate growth rates, but the SC group produced only a single subcutaneous nodule. The A group exhibited the highest tumour growth at 15 days post-surgery. Anatomic and histologic analyses corroborated the existence of various tumour nodules, and multiphoton microscopy was used to evaluate tumour fibrosis-infiltrating cells in a non-pathologic peritoneum. In conclusion, limited PM was obtained by IP injection, whereas IP injection after peritoneal scratching led to an extensive PM murine model for evaluating new therapeutics.
Collapse
Affiliation(s)
- Abdelkader Taibi
- Visceral Surgery Department, Dupuytren University Hospital, Limoges, France
- University Limoges, CNRS, XLIM, UMR 7252, Limoges, France
- * E-mail:
| | - Jeremie Albouys
- University Limoges, CNRS, XLIM, UMR 7252, Limoges, France
- Gastroenterology Department, Dupuytren University Hospital, Limoges, France
| | - Jeremie Jacques
- University Limoges, CNRS, XLIM, UMR 7252, Limoges, France
- Gastroenterology Department, Dupuytren University Hospital, Limoges, France
| | | | - Catherine Yardin
- University Limoges, CNRS, XLIM, UMR 7252, Limoges, France
- Cytology and Histology Department, Dupuytren University Hospital, Limoges, France
| | - Sylvaine Durand Fontanier
- Visceral Surgery Department, Dupuytren University Hospital, Limoges, France
- University Limoges, CNRS, XLIM, UMR 7252, Limoges, France
| | | |
Collapse
|
16
|
He Y, Gu Z, Zhu Q, Chen M, He C, Huang Y, Li Q, Di W. CA125 over‐release behavior following a 75‐g oral glucose test as a predictive biomarker of multidrug resistance in patients with ovarian cancer. Int J Cancer 2019; 145:1690-1700. [PMID: 30807642 DOI: 10.1002/ijc.32237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Yifeng He
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Tumor Microenvironment and Metastasis ProgramThe Wistar Institute, University of Pennsylvania Philadelphia PA
| | - Zhuowei Gu
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Qiujing Zhu
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Mo Chen
- Department of GynecologyObstetrics and Gynecology Hospital, Fudan University Shanghai China
| | - Chenghui He
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Yuting Huang
- Children's Research Institute, Children's National Medical Center Washington WA
| | - Qing Li
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- State Key Laboratory of Oncogene and Related GenesShanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Wen Di
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- State Key Laboratory of Oncogene and Related GenesShanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
17
|
Xu C, Li X, Liu P, Li M, Luo F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine. Oncol Lett 2019; 17:3-10. [PMID: 30655732 PMCID: PMC6313209 DOI: 10.3892/ol.2018.9583] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 05/16/2017] [Indexed: 12/31/2022] Open
Abstract
Patient-derived xenograft (PDX) mouse models involve the direct transfer of fresh human tumor samples into immunodeficient mice following surgical resection or other medical operations. Gene expression in tumors may be maintained by serial passages of tumors from mouse to mouse. These models aid research into tumor biology and pharmacology without manual manipulation of cell cultures in vitro. and are widely used in individualized cancer therapy/translational medicine, drug development and coclinical trials. PDX models exhibit higher predictive values for clinical outcomes than cell line-derived xenograft models and genetically engineered mouse models. However, PDX models are associated with certain challenges in clinical application. The present study reviewed current collections of PDX models and assessed the challenges and future directions of this field.
Collapse
Affiliation(s)
- Cong Xu
- Department of Acute Abdomen Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xuelu Li
- Department of Breast Surgery and Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Pixu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Man Li
- Department of Breast Surgery and Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Fuwen Luo
- Department of Acute Abdomen Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
18
|
Establishment of Patient-Derived Tumor Xenograft Models of High-Risk Endometrial Cancer. Int J Gynecol Cancer 2018; 28:1812-1820. [DOI: 10.1097/igc.0000000000001365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
ObjectiveHigh-risk endometrial cancers (ECs), including high-grade EC, serous carcinoma (SC), clear cell carcinoma, and carcinosarcoma, account for 50% of deaths due to ECs. Therapies for these cancers are limited, and patient-derived tumor xenograft (PDTX) models are useful tools for preclinical drug evaluation, biomarker identification, and personalized medicine strategies. Here, we used and compared 2 methods to establish PDTX models.MethodsFresh tumor tissues collected from 18 primary high-risk EC patients (10 high-grade ECs, 6 SCs, 1 clear cell carcinoma, and 1 carcinosarcoma) were engrafted subcutaneously and in the subrenal capsule in NOD/SCID for establishment and Balb/c-nu/nu mice for expansion. Histology and cytokeratin, estrogen receptor, progesterone receptor, and P53 expression were evaluated to assess the similarity of primary tumors and different generations of PDTX tumors. Whole-exome sequencing (WES) and RNA sequencing were used in 2 high-grade EC models to verify whether the genetic mutation profiles and gene expression were similar between primary and PDTX tumors.ResultsThe total tumor engraftment rate was 77.8% (14/18) regardless of the engraft method. The tumor engraftment rate was increased in subrenal capsule models compared with subcutaneous models (62.5% vs 50%, P = 0.464). The time to tumor formation varied significantly from 2 to 11 weeks. After subrenal capsular grafting, grafted tumors could be successfully transplanted to subcutaneous sites. We observed good similarity between primary tumors and corresponding different passages of xenografts.ConclusionsThe combination of 2 engrafting methods increases the tumor engraftment rate. The high tumor engraftment rate ensures the establishment of a high-risk EC biobank, which is a powerful resource for performing preclinical drug-sensitivity tests and identifying biomarkers for response or resistance.
Collapse
|
19
|
Lu W, Chao T, Ruiqi C, Juan S, Zhihong L. Patient-derived xenograft models in musculoskeletal malignancies. J Transl Med 2018; 16:107. [PMID: 29688859 PMCID: PMC5913806 DOI: 10.1186/s12967-018-1487-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022] Open
Abstract
Successful oncological drug development for bone and soft tissue sarcoma is grossly stagnating. A major obstacle in this process is the lack of appropriate animal models recapitulating the complexity and heterogeneity of musculoskeletal malignancies, resulting in poor efficiency in translating the findings of basic research to clinical applications. In recent years, patient-derived xenograft (PDX) models generated by directly engrafting patient-derived tumor fragments into immunocompromised mice have recaptured the attention of many researchers due to their properties of retaining the principle histopathology, biological behaviors, and molecular and genetic characteristics of the original tumor, showing promising future in both basic and clinical studies of bone and soft tissue sarcoma. Despite several limitations including low take rate and long take time in PDX generation, deficient immune system and heterologous tumor microenvironment of the host, PDXs offer a more advantageous platform for preclinical drug screening, biomarker identification and clinical therapeutic decision guiding. Here, we provide a timely review of the establishment and applications of PDX models for musculoskeletal malignancies and discuss current challenges and future directions of this approach.
Collapse
Affiliation(s)
- Wan Lu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Tu Chao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Chen Ruiqi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Su Juan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Li Zhihong
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China.
| |
Collapse
|
20
|
Li X, Zhu D, Li N, Yang H, Zhao Z, Li M. Characterization of ascites-derived tumor cells from an endometrial cancer patient. Cancer Sci 2017; 108:2352-2357. [PMID: 28945304 PMCID: PMC5715242 DOI: 10.1111/cas.13407] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022] Open
Abstract
Improved treatment outcomes for the endometrial cancer patient requires precision methods to investigate the biology of this disease and clinically relevant models to test treatment drugs. Hence, we applied a personalized platform to investigate whether in vitro and in vivo models could accurately predict effective treatment regimens. We successfully expanded ascites-derived tumor cells from an endometrial cancer patient with malignant ascites using ascites collected prior to chemotherapy treatment. Hematoxylin-eosin and immunohistochemistry staining of ascites-derived tumor cells confirmed the source of endometrial cancer cells. Ascites-derived tumor cells were sensitive to cisplatin and doxorubicin single-agent treatments in CCK-8 assay and 3-D culture, a condition that more closely mimics the in vivo environment. We further showed that ascites-derived tumor cells from this patient could form tumors in NOD/SCID mice with preserved morphological characteristics. A remarkable concordance between the clinical response of cisplatin and the results of in vitro and in vivo drug tests reflected the reliability of our personalized approach in this case. Together, our results indicated that an effective platform for ex vivo and in vivo culture of ascites-derived tumor cells from our endometrial cancer patient could be applied to identify treatment options, and may be commonly used in treating cancer patients with malignant ascites in the future.
Collapse
Affiliation(s)
- Xuelu Li
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
- Department of Critical Care MedicineThe Second Hospital of Dalian Medical UniversityDalianChina
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Dandan Zhu
- Department of Critical Care MedicineThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Ning Li
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Hui Yang
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Zuowei Zhao
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Man Li
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
21
|
Jung J, Kim J, Lim HK, Kim KM, Lee YS, Park JS, Yoon DS. Establishing a colorectal cancer liver metastasis patient-derived tumor xenograft model for the evaluation of personalized chemotherapy. Ann Surg Treat Res 2017; 93:173-180. [PMID: 29094026 PMCID: PMC5658298 DOI: 10.4174/astr.2017.93.4.173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/05/2017] [Accepted: 03/30/2017] [Indexed: 12/30/2022] Open
Abstract
Purpose In order to suggest optimal anticancer drugs for patient-tailored chemotherapy, we developed a colorectal cancer (CRC)-liver metastasis patient-derived tumor xenograft (PDTX) model. Methods Tissue obtained from a patient with CRC-liver metastasis (F0) was transplanted in a nonobese female mouse with diabetic/severe combined immune deficiency (F1) and the tumor tissue was retransplanted into nude mice (F2). When tumor volumes reached ~500 mm3, the F2 mice were randomly divided into 4 groups (n = 4/group) of doxorubicin, cisplatin, docetaxel, and nontreated control groups. The tumor tissues were investigated using H&E staining, terminal deoxynucleotidyl transferase dUTP nick end labeling assays, and immunohistochemistry. To determine where the mutant allele frequencies varied across the different passages, we isolated genomic DNA from the primary tumor, liver metastasis, and PDTX models (F1/F2). Results The physiological properties of the tumor were in accord with those of the patient's tumors. Anticancer drugs delayed tumor growth, inhibited proliferation, and caused apoptosis. Histological assessments revealed no observable heterogeneity among the intragenerational PDTX models. Target exon sequencing analysis without high-quality filter conditions revealed some genetic variations in the 83 cancer-related genes across the generations. However, when de novo mutations were defined as a total count of zero in F0 and ≥5 in F2, exactly prognostic impact of clone cancer profiling (EGFR, KRAS, BRAF, PIK3CA, NRAS, APC and TP53) were detected in the paired. Conclusion A CRC liver metastasis PDTX model was established for the evaluation of chemotherapeutic efficacy. This model retained the physiological characters of the patient tumors and potentially provides a powerful means of assessing chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Joohee Jung
- College of Pharmacy, Duksung Women's University, Seoul, Korea.,Innovative Drug Center, Duksung Women's University, Seoul, Korea
| | - Jisup Kim
- Department of Pathology, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Hyun Kyung Lim
- Innovative Drug Center, Duksung Women's University, Seoul, Korea
| | - Kyoung Mee Kim
- College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Yun Sun Lee
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Joon Seong Park
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Dong Sup Yoon
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| |
Collapse
|
22
|
Jung J, Seol HS, Chang S. The Generation and Application of Patient-Derived Xenograft Model for Cancer Research. Cancer Res Treat 2017; 50:1-10. [PMID: 28903551 PMCID: PMC5784646 DOI: 10.4143/crt.2017.307] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022] Open
Abstract
Establishing an appropriate preclinical model is crucial for translational cancer research. The most common way that has been adopted by far is grafting cancer cell lines, derived from patients. Although this xenograft model is easy to generate, but has several limitations because this cancer model could not represent the unique features of each cancer patient sufficiently. Moreover, accumulating evidences demonstrate cancer is a highly heterogeneous disease so that a tumor is comprised of cancer cells with diverse characteristics. In attempt to avoid these discrepancies between xenograft model and patients' tumor, a patient-derived xenograft (PDX) model has been actively generated and applied. The PDX model can be developed by the implantation of cancerous tissue from a patient's tumor into an immune-deficient mouse directly, thereby it preserves both cell-cell interactions and tumor microenvironment. In addition, the PDX model has shown advantages as a preclinical model in drug screening, biomarker development and co-clinical trial. In this review, we will summarize the methodology and applications of PDX in detail, and cover critical issues for the development of this model for preclinical research.
Collapse
Affiliation(s)
- Jaeyun Jung
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyang Sook Seol
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Suhwan Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Zervantonakis IK, Iavarone C, Chen HY, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis U, Leverson JD, Sampath D, Mills GB, Brugge JS. Systems analysis of apoptotic priming in ovarian cancer identifies vulnerabilities and predictors of drug response. Nat Commun 2017. [PMID: 28848242 DOI: 10.1038/s41467-017-00263-7]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2022] Open
Abstract
The lack of effective chemotherapies for high-grade serous ovarian cancers (HGS-OvCa) has motivated a search for alternative treatment strategies. Here, we present an unbiased systems-approach to interrogate a panel of 14 well-annotated HGS-OvCa patient-derived xenografts for sensitivity to PI3K and PI3K/mTOR inhibitors and uncover cell death vulnerabilities. Proteomic analysis reveals that PI3K/mTOR inhibition in HGS-OvCa patient-derived xenografts induces both pro-apoptotic and anti-apoptotic signaling responses that limit cell killing, but also primes cells for inhibitors of anti-apoptotic proteins. In-depth quantitative analysis of BCL-2 family proteins and other apoptotic regulators, together with computational modeling and selective anti-apoptotic protein inhibitors, uncovers new mechanistic details about apoptotic regulators that are predictive of drug sensitivity (BIM, caspase-3, BCL-XL) and resistance (MCL-1, XIAP). Our systems-approach presents a strategy for systematic analysis of the mechanisms that limit effective tumor cell killing and the identification of apoptotic vulnerabilities to overcome drug resistance in ovarian and other cancers.High-grade serous ovarian cancers (HGS-OvCa) frequently develop chemotherapy resistance. Here, the authors through a systematic analysis of proteomic and drug response data of 14 HGS-OvCa PDXs demonstrate that targeting apoptosis regulators can improve response of these tumors to inhibitors of the PI3K/mTOR pathway.
Collapse
Affiliation(s)
- Ioannis K Zervantonakis
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Claudia Iavarone
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Hsing-Yu Chen
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Laura M Selfors
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Sangeetha Palakurthi
- Belfer Center for Applied Cancer Research, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Joyce F Liu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Ursula Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Joel D Leverson
- Oncology Development, AbbVie, Inc, North Chicago, IL, 60064, USA
| | - Deepak Sampath
- Translational Oncology, Genentech, South San Francisco, CA, 94080, USA
| | - Gordon B Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joan S Brugge
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Systems analysis of apoptotic priming in ovarian cancer identifies vulnerabilities and predictors of drug response. Nat Commun 2017; 8:365. [PMID: 28848242 PMCID: PMC5573720 DOI: 10.1038/s41467-017-00263-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/15/2017] [Indexed: 12/15/2022] Open
Abstract
The lack of effective chemotherapies for high-grade serous ovarian cancers (HGS-OvCa) has motivated a search for alternative treatment strategies. Here, we present an unbiased systems-approach to interrogate a panel of 14 well-annotated HGS-OvCa patient-derived xenografts for sensitivity to PI3K and PI3K/mTOR inhibitors and uncover cell death vulnerabilities. Proteomic analysis reveals that PI3K/mTOR inhibition in HGS-OvCa patient-derived xenografts induces both pro-apoptotic and anti-apoptotic signaling responses that limit cell killing, but also primes cells for inhibitors of anti-apoptotic proteins. In-depth quantitative analysis of BCL-2 family proteins and other apoptotic regulators, together with computational modeling and selective anti-apoptotic protein inhibitors, uncovers new mechanistic details about apoptotic regulators that are predictive of drug sensitivity (BIM, caspase-3, BCL-XL) and resistance (MCL-1, XIAP). Our systems-approach presents a strategy for systematic analysis of the mechanisms that limit effective tumor cell killing and the identification of apoptotic vulnerabilities to overcome drug resistance in ovarian and other cancers. High-grade serous ovarian cancers (HGS-OvCa) frequently develop chemotherapy resistance. Here, the authors through a systematic analysis of proteomic and drug response data of 14 HGS-OvCa PDXs demonstrate that targeting apoptosis regulators can improve response of these tumors to inhibitors of the PI3K/mTOR pathway.
Collapse
|
25
|
Zervantonakis IK, Iavarone C, Chen HY, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis U, Leverson JD, Sampath D, Mills GB, Brugge JS. Systems analysis of apoptotic priming in ovarian cancer identifies vulnerabilities and predictors of drug response. Nat Commun 2017. [PMID: 28848242 DOI: 10.1038/s41467-017-00263-7] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The lack of effective chemotherapies for high-grade serous ovarian cancers (HGS-OvCa) has motivated a search for alternative treatment strategies. Here, we present an unbiased systems-approach to interrogate a panel of 14 well-annotated HGS-OvCa patient-derived xenografts for sensitivity to PI3K and PI3K/mTOR inhibitors and uncover cell death vulnerabilities. Proteomic analysis reveals that PI3K/mTOR inhibition in HGS-OvCa patient-derived xenografts induces both pro-apoptotic and anti-apoptotic signaling responses that limit cell killing, but also primes cells for inhibitors of anti-apoptotic proteins. In-depth quantitative analysis of BCL-2 family proteins and other apoptotic regulators, together with computational modeling and selective anti-apoptotic protein inhibitors, uncovers new mechanistic details about apoptotic regulators that are predictive of drug sensitivity (BIM, caspase-3, BCL-XL) and resistance (MCL-1, XIAP). Our systems-approach presents a strategy for systematic analysis of the mechanisms that limit effective tumor cell killing and the identification of apoptotic vulnerabilities to overcome drug resistance in ovarian and other cancers.High-grade serous ovarian cancers (HGS-OvCa) frequently develop chemotherapy resistance. Here, the authors through a systematic analysis of proteomic and drug response data of 14 HGS-OvCa PDXs demonstrate that targeting apoptosis regulators can improve response of these tumors to inhibitors of the PI3K/mTOR pathway.
Collapse
Affiliation(s)
- Ioannis K Zervantonakis
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Claudia Iavarone
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Hsing-Yu Chen
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Laura M Selfors
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Sangeetha Palakurthi
- Belfer Center for Applied Cancer Research, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Joyce F Liu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Ursula Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Joel D Leverson
- Oncology Development, AbbVie, Inc, North Chicago, IL, 60064, USA
| | - Deepak Sampath
- Translational Oncology, Genentech, South San Francisco, CA, 94080, USA
| | - Gordon B Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joan S Brugge
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Zhou R, Xu A, Gingold J, Strong LC, Zhao R, Lee DF. Li-Fraumeni Syndrome Disease Model: A Platform to Develop Precision Cancer Therapy Targeting Oncogenic p53. Trends Pharmacol Sci 2017; 38:908-927. [PMID: 28818333 DOI: 10.1016/j.tips.2017.07.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Li-Fraumeni syndrome (LFS) is a rare hereditary autosomal dominant cancer disorder. Germline mutations in TP53, the gene encoding p53, are responsible for most cases of LFS. TP53 is also the most commonly mutated gene in human cancers. Because inhibition of mutant p53 is considered to be a promising therapeutic strategy to treat these diseases, LFS provides a perfect genetic model to study p53 mutation-associated malignancies as well as to screen potential compounds targeting oncogenic p53. In this review we briefly summarize the biology of LFS and current understanding of the oncogenic functions of mutant p53 in cancer development. We discuss the strengths and limitations of current LFS disease models, and touch on existing compounds targeting oncogenic p53 and in vitro clinical trials to develop new ones. Finally, we discuss how recently developed methodologies can be integrated into the LFS induced pluripotent stem cell (iPSC) platform to develop precision cancer therapy.
Collapse
Affiliation(s)
- Ruoji Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; These authors contributed equally to this work
| | - An Xu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; These authors contributed equally to this work
| | - Julian Gingold
- Women's Health Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; These authors contributed equally to this work
| | - Louise C Strong
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ruiying Zhao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Dickreuter E, Cordes N. The cancer cell adhesion resistome: mechanisms, targeting and translational approaches. Biol Chem 2017; 398:721-735. [PMID: 28002024 DOI: 10.1515/hsz-2016-0326] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
Abstract
Cell adhesion-mediated resistance limits the success of cancer therapies and is a great obstacle to overcome in the clinic. Since the 1990s, where it became clear that adhesion of tumor cells to the extracellular matrix is an important mediator of therapy resistance, a lot of work has been conducted to understand the fundamental underlying mechanisms and two paradigms were deduced: cell adhesion-mediated radioresistance (CAM-RR) and cell adhesion-mediated drug resistance (CAM-DR). Preclinical work has evidently demonstrated that targeting of integrins, adapter proteins and associated kinases comprising the cell adhesion resistome is a promising strategy to sensitize cancer cells to both radiotherapy and chemotherapy. Moreover, the cell adhesion resistome fundamentally contributes to adaptation mechanisms induced by radiochemotherapy as well as molecular drugs to secure a balanced homeostasis of cancer cells for survival and growth. Intriguingly, this phenomenon provides a basis for synthetic lethal targeted therapies simultaneously administered to standard radiochemotherapy. In this review, we summarize current knowledge about the cell adhesion resistome and highlight targeting strategies to override CAM-RR and CAM-DR.
Collapse
Affiliation(s)
| | - Nils Cordes
- , Faculty of Medicine and University Hospital Carl Gustav Carus
| |
Collapse
|
28
|
Szadvari I, Krizanova O, Babula P. Athymic nude mice as an experimental model for cancer treatment. Physiol Res 2017; 65:S441-S453. [PMID: 28006926 DOI: 10.33549/physiolres.933526] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Athymic nude mice, a murine strain bearing spontaneous deletion in the Foxn1 gene that causes deteriorated or absent thymus (which results in inhibited immune system with reduction of number of T cells), represent a widely used model in cancer research having long lasting history as a tool for preclinical testing of drugs. The review describes three models of athymic mice that utilize cancer cell lines to induce tumors. In addition, various methods that can be applied in order to evaluate activity of anticancer agents in these models are shown and discussed. Although each model has certain disadvantages, they are still considered as inevitable instruments in many fields of cancer research, particularly in finding new drugs that would more effectively combat the cancer disease or enhance the use of current chemotherapy. Finally, the review summarizes strengths and weaknesses as well as future perspectives of the athymic nude mice model in cancer research.
Collapse
Affiliation(s)
- I Szadvari
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | | | |
Collapse
|
29
|
Liu X, Krawczyk E, Suprynowicz FA, Palechor-Ceron N, Yuan H, Dakic A, Simic V, Zheng YL, Sripadhan P, Chen C, Lu J, Hou TW, Choudhury S, Kallakury B, Tang DG, Darling T, Thangapazham R, Timofeeva O, Dritschilo A, Randell SH, Albanese C, Agarwal S, Schlegel R. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat Protoc 2017; 12:439-451. [PMID: 28125105 PMCID: PMC6195120 DOI: 10.1038/nprot.2016.174] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Historically, it has been difficult to propagate cells in vitro that are derived directly from human tumors or healthy tissue. However, in vitro preclinical models are essential tools for both the study of basic cancer biology and the promotion of translational research, including drug discovery and drug target identification. This protocol describes conditional reprogramming (CR), which involves coculture of irradiated mouse fibroblast feeder cells with normal and tumor human epithelial cells in the presence of a Rho kinase inhibitor (Y-27632). CR cells can be used for various applications, including regenerative medicine, drug sensitivity testing, gene expression profiling and xenograft studies. The method requires a pathologist to differentiate healthy tissue from tumor tissue, and basic tissue culture skills. The protocol can be used with cells derived from both fresh and cryopreserved tissue samples. As approximately 1 million cells can be generated in 7 d, the technique is directly applicable to diagnostic and predictive medicine. Moreover, the epithelial cells can be propagated indefinitely in vitro, yet retain the capacity to become fully differentiated when placed into conditions that mimic their natural environment.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Correspondence should be addressed to X.L. () or R.S. ()
| | - Ewa Krawczyk
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Correspondence should be addressed to X.L. () or R.S. ()
| | - Frank A Suprynowicz
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Nancy Palechor-Ceron
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Hang Yuan
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Aleksandra Dakic
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Vera Simic
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Yun-Ling Zheng
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Praathibha Sripadhan
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Chen Chen
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Jie Lu
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Tung-Wei Hou
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Sujata Choudhury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Bhaskar Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Thomas Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Rajesh Thangapazham
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Olga Timofeeva
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Anatoly Dritschilo
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Christopher Albanese
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Seema Agarwal
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
30
|
Heo EJ, Cho YJ, Cho WC, Hong JE, Jeon HK, Oh DY, Choi YL, Song SY, Choi JJ, Bae DS, Lee YY, Choi CH, Kim TJ, Park WY, Kim BG, Lee JW. Patient-Derived Xenograft Models of Epithelial Ovarian Cancer for Preclinical Studies. Cancer Res Treat 2017; 49:915-926. [PMID: 28052650 PMCID: PMC5654149 DOI: 10.4143/crt.2016.322] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022] Open
Abstract
Purpose Patient-derived tumor xenografts (PDXs) can provide more reliable information about tumor biology than cell line models. We developed PDXs for epithelial ovarian cancer (EOC) that have histopathologic and genetic similarities to the primary patient tissues and evaluated their potential for use as a platform for translational EOC research. Materials and Methods We successfully established PDXs by subrenal capsule implantation of primary EOC tissues into female BALB/C-nude mice. The rate of successful PDX engraftment was 48.8% (22/45 cases). Hematoxylin and eosin staining and short tandem repeat analysis showed histopathological and genetic similarity between the PDX and primary patient tissues. Results Patients whose tumors were successfully engrafted in mice had significantly inferior overall survival when compared with those whose tumors failed to engraft (p=0.040). In preclinical tests of this model, we found that paclitaxel-carboplatin combination chemotherapy significantly deceased tumor weight in PDXs compared with the control treatment (p=0.013). Moreover, erlotinib treatment significantly decreased tumor weight in epidermal growth factor receptor–overexpressing PDX with clear cell histology (p=0.023). Conclusion PDXs for EOC with histopathological and genetic stability can be efficiently developed by subrenal capsule implantation and have the potential to provide a promising platform for future translational research and precision medicine for EOC.
Collapse
Affiliation(s)
- Eun Jin Heo
- Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Young Jae Cho
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - William Chi Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Ji Eun Hong
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye-Kyung Jeon
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Doo-Yi Oh
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoo-Young Lee
- Division of Gynecologic Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae-Joong Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea.,Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Chaudary N, Pintilie M, Hedley D, Hill RP, Milosevic M, Mackay H. Hedgehog inhibition enhances efficacy of radiation and cisplatin in orthotopic cervical cancer xenografts. Br J Cancer 2016; 116:50-57. [PMID: 27875522 PMCID: PMC5220149 DOI: 10.1038/bjc.2016.383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/07/2016] [Accepted: 10/21/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The Hedgehog (Hh) pathway is upregulated in cervical cancer and associated with poor outcome. We explored the effects of Hh pathway inhibition in combination with RTCT in a patient derived orthotopic cervical cancer xenograft model (OCICx). METHODS 5E1, a monoclonal antibody for SHH, or Sonidegib (LDE225), a clinical SMO inhibitor (Novartis) were added to RTCT. We investigated tumour growth delay, metastasis and GI toxicity using orthotopic cervical cancer xenografts models. The xenografts were treated with radiotherapy (15 × 2 Gy daily fractions over 3 weeks) and weekly cisplatin 4 mg kg-1 concurrently, with or without 5E1 or Sonidegib (LDE225). The Hh inhibitors were administered by subcutaneous injection (5E1; 20 mg kg-1 weekly for 3 weeks), or by oral gavage (Sonidegib; 60 mg kg-1 daily for 3 weeks). RESULTS We observed that both Hh inhibitors administered with RTCT were well tolerated and showed increased tumour growth delay, and reduced metastasis, with no increase in acute GI-toxicity relative to RTCT alone. CONCLUSIONS Our data suggest Hh can be a valid therapeutic target in cervical cancer and supports data suggesting a potential therapeutic role for targeting Hh in patients undergoing RTCT. This warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Naz Chaudary
- Ontario Cancer Institute/Princess Margaret Cancer Centre and The Campbell Family Institute for Cancer Research, Ontario MG5 2M9, Canada
| | - Melania Pintilie
- Biostatistics Department, Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario MG5 2M9, Canada.,Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario M5T 3M7, Canada
| | - David Hedley
- Ontario Cancer Institute/Princess Margaret Cancer Centre and The Campbell Family Institute for Cancer Research, Ontario MG5 2M9, Canada.,Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario MG5 2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario MG5 2M9, Canada
| | - Richard P Hill
- Ontario Cancer Institute/Princess Margaret Cancer Centre and The Campbell Family Institute for Cancer Research, Ontario MG5 2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario MG5 2M9, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario MG5 2M9, Canada
| | - Michael Milosevic
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario MG5 2M9, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario MG5 2M9, Canada
| | - Helen Mackay
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario MG5 2M9, Canada
| |
Collapse
|
32
|
Abstract
Tumours contain multiple different cell populations, including cells derived from the bone marrow as well as cancer-associated fibroblasts and various stromal populations including the vasculature. The microenvironment of the tumour cells plays a significant role in the response of the tumour to radiation treatment. Low levels of oxygen (hypoxia) caused by the poorly organized vasculature in tumours have long been known to affect radiation response; however, other aspects of the microenvironment may also play important roles. This article reviews some of the old literature concerning tumour response to irradiation and relates this to current concepts about the role of the tumour microenvironment in tumour response to radiation treatment. Included in the discussion are the role of cancer stem cells, radiation damage to the vasculature and the potential for radiation to enhance immune activity against tumour cells. Radiation treatment can cause a significant influx of bone marrow-derived cell populations into both normal tissues and tumours. Potential roles of such cells may include enhancing vascular recovery as well as modulating immune reactivity.
Collapse
Affiliation(s)
- Richard P Hill
- 1 Ontario Cancer Institute, Princess Margaret Cancer Centre, Toronto, ON, Canada.,2 Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Tabb DL, Wang X, Carr SA, Clauser KR, Mertins P, Chambers MC, Holman JD, Wang J, Zhang B, Zimmerman LJ, Chen X, Gunawardena HP, Davies SR, Ellis MJC, Li S, Townsend RR, Boja ES, Ketchum KA, Kinsinger CR, Mesri M, Rodriguez H, Liu T, Kim S, McDermott JE, Payne SH, Petyuk VA, Rodland KD, Smith RD, Yang F, Chan DW, Zhang B, Zhang H, Zhang Z, Zhou JY, Liebler DC. Reproducibility of Differential Proteomic Technologies in CPTAC Fractionated Xenografts. J Proteome Res 2015; 15:691-706. [PMID: 26653538 PMCID: PMC4779376 DOI: 10.1021/acs.jproteome.5b00859] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The NCI Clinical Proteomic Tumor Analysis Consortium (CPTAC) employed a pair of reference xenograft proteomes for initial platform validation and ongoing quality control of its data collection for The Cancer Genome Atlas (TCGA) tumors. These two xenografts, representing basal and luminal-B human breast cancer, were fractionated and analyzed on six mass spectrometers in a total of 46 replicates divided between iTRAQ and label-free technologies, spanning a total of 1095 LC-MS/MS experiments. These data represent a unique opportunity to evaluate the stability of proteomic differentiation by mass spectrometry over many months of time for individual instruments or across instruments running dissimilar workflows. We evaluated iTRAQ reporter ions, label-free spectral counts, and label-free extracted ion chromatograms as strategies for data interpretation (source code is available from http://homepages.uc.edu/~wang2x7/Research.htm ). From these assessments, we found that differential genes from a single replicate were confirmed by other replicates on the same instrument from 61 to 93% of the time. When comparing across different instruments and quantitative technologies, using multiple replicates, differential genes were reproduced by other data sets from 67 to 99% of the time. Projecting gene differences to biological pathways and networks increased the degree of similarity. These overlaps send an encouraging message about the maturity of technologies for proteomic differentiation.
Collapse
Affiliation(s)
| | - Xia Wang
- Department of Mathematical Sciences, University of Cincinnati , Cincinnati, Ohio 45221, United States
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard , Cambridge, Massachusetts 02142, United States
| | - Karl R Clauser
- Proteomics Platform, Broad Institute of MIT and Harvard , Cambridge, Massachusetts 02142, United States
| | - Philipp Mertins
- Proteomics Platform, Broad Institute of MIT and Harvard , Cambridge, Massachusetts 02142, United States
| | | | | | | | | | | | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Harsha P Gunawardena
- Department of Biochemistry and Biophysics, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Sherri R Davies
- Department of Medicine, Washington University , St. Louis, Missouri 63110, United States
| | - Matthew J C Ellis
- Department of Medicine, Washington University , St. Louis, Missouri 63110, United States
| | - Shunqiang Li
- Department of Medicine, Washington University , St. Louis, Missouri 63110, United States
| | - R Reid Townsend
- Department of Medicine, Washington University , St. Louis, Missouri 63110, United States
| | - Emily S Boja
- Office of Cancer Clinical Proteomics Research, National Cancer Institute , Bethesda, Maryland 20892, United States
| | - Karen A Ketchum
- Enterprise Science and Computing, Inc. , Rockville, Maryland 20850, United States
| | - Christopher R Kinsinger
- Office of Cancer Clinical Proteomics Research, National Cancer Institute , Bethesda, Maryland 20892, United States
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute , Bethesda, Maryland 20892, United States
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute , Bethesda, Maryland 20892, United States
| | - Tao Liu
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Sangtae Kim
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Jason E McDermott
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Samuel H Payne
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Vladislav A Petyuk
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Karin D Rodland
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Richard D Smith
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Feng Yang
- Division of Biological Sciences, Pacific Northwest National Laboratory , Richland, Washington 99352, United States
| | - Daniel W Chan
- JHMI and Division of Clinical Chemistry, Johns Hopkins University , Baltimore, Maryland 21231, United States
| | - Bai Zhang
- JHMI and Division of Clinical Chemistry, Johns Hopkins University , Baltimore, Maryland 21231, United States
| | - Hui Zhang
- JHMI and Division of Clinical Chemistry, Johns Hopkins University , Baltimore, Maryland 21231, United States
| | - Zhen Zhang
- JHMI and Division of Clinical Chemistry, Johns Hopkins University , Baltimore, Maryland 21231, United States
| | - Jian-Ying Zhou
- JHMI and Division of Clinical Chemistry, Johns Hopkins University , Baltimore, Maryland 21231, United States
| | | |
Collapse
|