1
|
Koga Y, Ochiai A. Systematic Review of Patient-Derived Xenograft Models for Preclinical Studies of Anti-Cancer Drugs in Solid Tumors. Cells 2019; 8:cells8050418. [PMID: 31064068 PMCID: PMC6562882 DOI: 10.3390/cells8050418] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/26/2019] [Accepted: 05/04/2019] [Indexed: 01/06/2023] Open
Abstract
Patient-derived xenograft (PDX) models are used as powerful tools for understanding cancer biology in PDX clinical trials and co-clinical trials. In this systematic review, we focus on PDX clinical trials or co-clinical trials for drug development in solid tumors and summarize the utility of PDX models in the development of anti-cancer drugs, as well as the challenges involved in this approach, following the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Recently, the assessment of drug efficacy by PDX clinical and co-clinical trials has become an important method. PDX clinical trials can be used for the development of anti-cancer drugs before clinical trials, with their efficacy assessed by the modified response evaluation criteria in solid tumors (mRECIST). A few dozen cases of PDX models have completed enrollment, and the efficacy of the drugs is assessed by 1 × 1 × 1 or 3 × 1 × 1 approaches in the PDX clinical trials. Furthermore, co-clinical trials can be used for personalized care or precision medicine with the evaluation of a new drug or a novel combination. Several PDX models from patients in clinical trials have been used to assess the efficacy of individual drugs or drug combinations in co-clinical trials.
Collapse
Affiliation(s)
- Yoshikatsu Koga
- Department of Strategic Programs, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.
| | - Atsushi Ochiai
- Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.
| |
Collapse
|
2
|
Abstract
Immunotherapy is one of the most exciting recent breakthroughs in the field of cancer treatment. Many different approaches are being developed and a number have already gained regulatory approval or are under investigation in clinical trials. However, learning from the past, preclinical animal models often insufficiently reflect the physiological situation in humans, which subsequently causes treatment failures in clinical trials. Due to species-specific differences in most parts of the immune system, the transfer of knowledge from preclinical studies to clinical trials is eminently challenging. Human tumor cell line-based or patient-derived xenografts in immunocompromised mice have been successfully applied in the preclinical testing of cytotoxic or molecularly targeted agents, but naturally these systems lack the human immune system counterpart. The co-transplantation of human peripheral blood mononuclear cells or hematopoietic stem cells is employed to overcome this limitation. This review summarizes some important aspects of the different available tumor xenograft mouse models, their history, and their implementation in drug development and personalized therapy. Moreover, recent progress, opportunities and limitations of different humanized mouse models will be discussed.
Collapse
|
3
|
Synthesis & antitumor activity of epothilones B and D and their analogs. Future Med Chem 2018; 10:1483-1496. [DOI: 10.4155/fmc-2017-0320] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Epothilone is a newly developed antitumor drug; its antitumor principle is to stop the cell cycle by binding to tubulin in tumor cells, promoting tubulin polymerization, inhibiting depolymerization of microtubules, and ultimately inducing apoptosis. There are many analogs of epothilone, such as epothilone B, epothilone D, ixabepilone, sagopilone, 21-amino-epothilone B and KOS-1584. Herein, the synthesis and antitumor activity of epothilones B and D were summed up. The antitumor activity of epothilone analogs was also compared. Synthesis of epothilone and its analogs is more complex, and choosing the proper synthetic method is very important. Moreover, these compounds have obvious antitumor effect. The epothilone and its analogs will continue to play an important role in the future treatment of tumors.
Collapse
|
4
|
Abstract
Mutations in cancer cells frequently result in cell cycle alterations that lead to unrestricted growth compared to normal cells. Considering this phenomenon, many drugs have been developed to inhibit different cell-cycle phases. Mitotic phase targeting disturbs mitosis in tumor cells, triggers the spindle assembly checkpoint and frequently results in cell death. The first anti-mitotics to enter clinical trials aimed to target tubulin. Although these drugs improved the treatment of certain cancers, and many anti-microtubule compounds are already approved for clinical use, severe adverse events such as neuropathies were observed. Since then, efforts have been focused on the development of drugs that also target kinases, motor proteins and multi-protein complexes involved in mitosis. In this review, we summarize the major proteins involved in the mitotic phase that can also be targeted for cancer treatment. Finally, we address the activity of anti-mitotic drugs tested in clinical trials in recent years.
Collapse
|
5
|
Abstract
Experimental oncology research and preclinical drug development both substantially require specific, clinically relevant in vitro and in vivo tumor models. The increasing knowledge about the heterogeneity of cancer requested a substantial restructuring of the test systems for the different stages of development. To be able to cope with the complexity of the disease, larger panels of patient-derived tumor models have to be implemented and extensively characterized. Together with individual genetically engineered tumor models and supported by core functions for expression profiling and data analysis, an integrated discovery process has been generated for predictive and personalized drug development.Improved “humanized” mouse models should help to overcome current limitations given by xenogeneic barrier between humans and mice. Establishment of a functional human immune system and a corresponding human microenvironment in laboratory animals will strongly support further research.Drug discovery, systems biology, and translational research are moving closer together to address all the new hallmarks of cancer, increase the success rate of drug development, and increase the predictive value of preclinical models.
Collapse
|
6
|
Li J, Ye C, Mansmann UR. Comparing Patient-Derived Xenograft and Computational Response Prediction for Targeted Therapy in Patients of Early-Stage Large Cell Lung Cancer. Clin Cancer Res 2016; 22:2167-76. [PMID: 26637276 DOI: 10.1158/1078-0432.ccr-15-2401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Targeted therapy (TT) provides highly effective cancer treatment for appropriately selected individuals. A major challenge of TT is to select patients who would benefit most. EXPERIMENTAL DESIGN The study uses cancer material from 25 patients primarily diagnosed with non-small cell lung cancer (NSCLC). Patient-derived xenografts (PDXs) are treated with cetuximab and erlotinib. Treatment response is measured by tumor shrinkage comparing tumor volume at day 25 (V25) with tumor volume at baseline (V0). Shrinkage below 40% is considered as treatment response: V25/V0 < 0.4 (<40%). Furthermore, RNA-seq data from each tumor sample are used to predict tumor response to either treatment using an in silico molecular signaling map (MSM) approach. RESULTS PDX response was 40% (10/25; 95% CI [21.13%, 61.34%]) under cetuximab and 20% (5/25; 95% CI [6.83%, 40.70%]) under erlotinib. MSM predicted response was 48% (12/25; 95% CI [27.8%, 68.7%]) under cetuximab and 40% (10/25; 95% CI [21.13%, 61.34%]) under erlotinib. Agreement between PDX and MSM response prediction is substantial under cetuximab and erlotinib: 84% (21/25, P = 0.001) and 80% (20/25, P = 0.003). A total of 5 from the 25 patients have been treated with cetuximab showing a clinical response identical to both predictions. CONCLUSIONS For NSCLC patients, this proof-of-concept study shows a considerable agreement in response prediction from MSM and PDX approaches, but MSM saves time and laboratory resources. Our result indicates the potential of MSM-based approach for clinical decision making when selecting cancer TTs. Clin Cancer Res; 22(9); 2167-76. ©2015 AACR.
Collapse
Affiliation(s)
- Jian Li
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Changkun Ye
- Medical Research Center of Yu Huang Hospital, Yu Huang, Zhejiang, PR China
| | - Ulrich R Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
7
|
Rogalska A, Sliwinska A, Kasznicki J, Drzewoski J, Marczak A. Effects of Epothilone A in Combination with the Antidiabetic Drugs Metformin and Sitagliptin in HepG2 Human Hepatocellular Cancer Cells: Role of Transcriptional Factors NF-κB and p53. Asian Pac J Cancer Prev 2016; 17:993-1001. [DOI: 10.7314/apjcp.2016.17.3.993] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
8
|
Chen WC, Yuan JS, Xing Y, Mitchell A, Mbong N, Popescu AC, McLeod J, Gerhard G, Kennedy JA, Bogdanoski G, Lauriault S, Perdu S, Merkulova Y, Minden MD, Hogge DE, Guidos C, Dick JE, Wang JCY. An Integrated Analysis of Heterogeneous Drug Responses in Acute Myeloid Leukemia That Enables the Discovery of Predictive Biomarkers. Cancer Res 2016; 76:1214-24. [PMID: 26833125 DOI: 10.1158/0008-5472.can-15-2743] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/17/2015] [Indexed: 11/16/2022]
Abstract
Many promising new cancer drugs proceed through preclinical testing and early-phase trials only to fail in late-stage clinical testing. Thus, improved models that better predict survival outcomes and enable the development of biomarkers are needed to identify patients most likely to respond to and benefit from therapy. Here, we describe a comprehensive approach in which we incorporated biobanking, xenografting, and multiplexed phospho-flow (PF) cytometric profiling to study drug response and identify predictive biomarkers in acute myeloid leukemia (AML) patients. To test the efficacy of our approach, we evaluated the investigational JAK2 inhibitor fedratinib (FED) in 64 patient samples. FED robustly reduced leukemia in mouse xenograft models in 59% of cases and was also effective in limiting the protumorigenic activity of leukemia stem cells as shown by serial transplantation assays. In parallel, PF profiling identified FED-mediated reduction in phospho-STAT5 (pSTAT5) levels as a predictive biomarker of in vivo drug response with high specificity (92%) and strong positive predictive value (93%). Unexpectedly, another JAK inhibitor, ruxolitinib (RUX), was ineffective in 8 of 10 FED-responsive samples. Notably, this outcome could be predicted by the status of pSTAT5 signaling, which was unaffected by RUX treatment. Consistent with this observed discrepancy, PF analysis revealed that FED exerted its effects through multiple JAK2-independent mechanisms. Collectively, this work establishes an integrated approach for testing novel anticancer agents that captures the inherent variability of response caused by disease heterogeneity and in parallel, facilitates the identification of predictive biomarkers that can help stratify patients into appropriate clinical trials.
Collapse
Affiliation(s)
- Weihsu C Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Julie S Yuan
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Yan Xing
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nathan Mbong
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andreea C Popescu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jessica McLeod
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gitte Gerhard
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Goce Bogdanoski
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Stevan Lauriault
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sofie Perdu
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yulia Merkulova
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada. Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Donna E Hogge
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Cynthia Guidos
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada. Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Cui YQ, Geng Q, Yu T, Zhang FL, Lin HC, Li J, Zhu MX, Liu L, Yao M, Yan MX. Establishment of a highly metastatic model with a newly isolated lung adenocarcinoma cell line. Int J Oncol 2015; 47:927-40. [PMID: 26134302 DOI: 10.3892/ijo.2015.3065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/18/2015] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of malignancy-related death worldwide, and metastasis always results in a poor prognosis. However, therapeutic progress is hampered by a deficiency of appropriate pre-clinical metastatic models. To bridge this experimental gap, we developed an in vivo metastatic model via subcutaneous (s.c.) injection. The original cell line (XL-2) adopted in this model was newly isolated from the ascites of a patient with extensive metastases of lung adenocarcinoma, thereby avoiding any alteration of its initial molecular biology features from artificial serial cultivation. After comprehensive phenotypical and histological analysis, it was identified as a lung adenocarcinoma cell line. Additionally, the drug test showed that XL-2 cell line was sensitive to docetaxel, and resistant to doxorubicin, indicating it might serve as a cell line model of drug resistance for identifying mechanisms of tumors resistant to doxorubicin. Through this s.c. model, we further obtained a highly metastatic cell line (designated XL-2sci). The metastatic rate of mice in XL-2 group was 3/10, in contrast to the rate of 9/10 in XL-2sci group. Optical imaging, micro-computed tomography (micro-CT) scanning and Transwell assays were further applied to identify the enhanced metastatic capacity of Xl-2sci cells both in vivo and in vitro. Compared with XL-2 cells, ITRAQ labeled proteomics profiling study showed that some tumor metastasis-associated proteins were upregulated in XL-2sci cells, which also indicated the reliability of our model. Proliferation ability of XL-2 and XL-2sci were also evaluated. Results showed that highly metastatic XL-2sci possessed a decreased proliferation capacity versus XL-2, which demonstrated that its increased metastatic activity was not facilitated by a faster growth rate. In conclusion, we successfully developed an in vivo metastatic model using a newly established lung adenocarcinoma cell line, which will be beneficial to further investigations of lung cancer metastasis and to the development of anti-metastasis drugs.
Collapse
Affiliation(s)
- Yong-Qi Cui
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Qin Geng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Tao Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Fang-Lin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - He-Chun Lin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Jing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Miao-Xin Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Lei Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Ming-Xia Yan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| |
Collapse
|
10
|
Abstract
Patient-derived xenograft (PDX) models are now being widely used in cancer research and have the potential to greatly inform our understanding of cancer biology. However, many questions remain, especially regarding the ability of PDX models to affect clinical decision making. With these points in mind, we asked three scientists to give their opinions on the generation and uses of PDX models and the future of this field.
Collapse
Affiliation(s)
- Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1L3, British Columbia, Canada
| | - Manuel Hidalgo
- Gastrointestinal Cancer Clinical Research Unit, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Andrew L Kung
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
11
|
Russo MV, Faversani A, Gatti S, Ricca D, Del Gobbo A, Ferrero S, Palleschi A, Vaira V, Bosari S. A new mouse avatar model of non-small cell lung cancer. Front Oncol 2015; 5:52. [PMID: 25785245 PMCID: PMC4347595 DOI: 10.3389/fonc.2015.00052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/13/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction: Lung cancer remains the leading cause of tumor-related deaths, despite advances in the understanding of the disease pathogenesis and in its clinical treatment. It is crucial to develop novel technologies to discover disease biomarkers and predict individual therapy response. Materials and methods: We established 48 patients-derived tumor xenografts (PDTXs) implanted in the subrenal capsule of immunodeficient mice using thin, precision-cut tumor tissue slices, derived from five patients affected by non-small cell lung cancer. Twenty-six tissue slices were immediately processed and implanted at sample recovery [patients-derived tumor xenografts derived from fresh tissue (dPDTX)], whereas the remaining sections were cultured on specific organotypic supports at 37°C and 5% CO2 for 24 h before grafting [patients-derived tumor xenografts derived from cultured tissue (cPDTX)]. At sacrifice, xenografts tissue morphology, proliferation (Ki67), and histotype markers were analyzed. Oncogenic miRNAs profiles were assessed in PDTXs, human tumors, and serum from one patient. Results: Xenografts retained the original cancer features and there were no differences between dPDTXs and cPDTXs. Squamous cell carcinoma (SCC) xenografts showed a higher engraftment rate than adenocarcinoma (AC)-derived tumors. At basal time, Ki67 levels were higher in SCCs than in ACs, and the expression levels of genes associated to a stem cell-like phenotype were also more expressed in SCC samples. The analysis of oncogenic miRNAs showed that circulating miR-19b, -21, and -210 levels were correlated with higher Ki67 expression in xenografts. Conclusion: Our study implemented the PDTX model with thin, precision-cut tumor slices from small tumors, which could be useful for clinical applications and predictive purposes. The different engraftment success is likely determined by tumor histotype, high proliferation index, and the expression of genes essential for cancer stem cells maintenance. Our PDTXs model could be a valid tool to expand primary tumors for the discovery of new biomarkers and explore therapeutic options.
Collapse
Affiliation(s)
- Maria Veronica Russo
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy ; Department of Pathophysiology and Transplantation, Doctorate School in Molecular and Translational Medicine, University of Milan , Milan , Italy
| | - Alice Faversani
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Stefano Gatti
- Center for Preclinical Surgical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy ; Department of Medical Biotechnology and Translational Medicine, University of Milan , Milan , Italy
| | - Dario Ricca
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Alessandro Del Gobbo
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy ; Department of Biomedical, Surgical and Dental Sciences, University of Milan , Milan , Italy
| | - Alessandro Palleschi
- Division of Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy ; Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi" (INGM) , Milan , Italy
| | - Silvano Bosari
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy ; Department of Pathophysiology and Transplantation, Doctorate School in Molecular and Translational Medicine, University of Milan , Milan , Italy
| |
Collapse
|
12
|
Ilie M, Nunes M, Blot L, Hofman V, Long-Mira E, Butori C, Selva E, Merino-Trigo A, Vénissac N, Mouroux J, Vrignaud P, Hofman P. Setting up a wide panel of patient-derived tumor xenografts of non-small cell lung cancer by improving the preanalytical steps. Cancer Med 2014; 4:201-11. [PMID: 25470237 PMCID: PMC4329004 DOI: 10.1002/cam4.357] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/26/2014] [Accepted: 08/31/2014] [Indexed: 12/30/2022] Open
Abstract
With the ongoing need to improve therapy for non–small cell lung cancer (NSCLC) there has been increasing interest in developing reliable preclinical models to test novel therapeutics. Patient-derived tumor xenografts (PDX) are considered to be interesting candidates. However, the establishment of such model systems requires highly specialized research facilities and introduces logistic challenges. We aimed to establish an extensive well-characterized panel of NSCLC xenograft models in the context of a long-distance research network after careful control of the preanalytical steps. One hundred fresh surgically resected NSCLC specimens were shipped in survival medium at room temperature from a hospital-integrated biobank to animal facilities. Within 24 h post-surgery, tumor fragments were subcutaneously xenografted into immunodeficient mice. PDX characterization was performed by histopathological, immunohistochemical, aCGH and next-generation sequencing approaches. For this model system, the tumor take rate was 35%, with higher rates for squamous carcinoma (60%) than for adenocarcinoma (13%). Patients for whom PDX tumors were obtained had a significantly shorter disease-free survival (DFS) compared to patients for whom no PDX tumors (P = 0.039) were obtained. We established a large panel of PDX NSCLC models with a high frequency of mutations (29%) in EGFR, KRAS, NRAS, MEK1, BRAF, PTEN, and PI3KCA genes and with gene amplification (20%) of c-MET and FGFR1. This new patient-derived NSCLC xenograft collection, established regardless of the considerable time required and the distance between the clinic and the animal facilities, recapitulated the histopathology and molecular diversity of NSCLC and provides stable and reliable preclinical models for human lung cancer research.
Collapse
Affiliation(s)
- Marius Ilie
- Laboratory of Clinical and Experimental Pathology, Louis Pasteur Hospital, Nice, France; IRCAN Team 3, Inserm U1081/UMR CNRS 7284, Faculty of Medicine of Nice, University of Nice Sophia Antipolis, Nice, France; Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France; Hospital Related Biobank BB-0033-00025, Louis Pasteur Hospital, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Lee WS, Kim HY, Seok JY, Jang HH, Park YH, Kim SY, Shin DB, Hong S. Genomic profiling of patient-derived colon cancer xenograft models. Medicine (Baltimore) 2014; 93:e298. [PMID: 25526474 PMCID: PMC4603094 DOI: 10.1097/md.0000000000000298] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent evidence suggests that patient derived xenograft (PDX) models can maintain certain pathological and molecular features of the original disease. However, these characterizations are limited to immunohistochemistry or by tissue microarray analysis. We conducted a high-throughput sequencing of primary colon tumor and PDX has not been reported yet. Fresh primary colon cancer tissues that originate from surgery were implanted into the subcutaneous space of 6- to 8-week-old female BALB/c nu/nu or NOD/SCID mice and serially passaged in vivo. Ion AmpliSeq Cancer Hotspot Panel v2 (Ion Torrent) was used to detect frequent somatic mutations and similarity of molecular characteristics between the 10 patient tumors and matched PDX. Histologic and immunohistochemical analyses revealed a high degree of pathologic similarity including histologic architecture and expression of CEA, CK7, and CD20 between the patient and xenograft tumors. In 80% cases, all of the somatic mutations detected in primary tumor were concordantly detected in PDX models. However, 2 PDX models showed gained mutations such as PIK3CA or FBWX7 mutation. Ten patient-derived advanced colon cancer xenograft models were established. These models maintained the key characteristic features of the original tumors, suggesting useful tool for preclinical personalized medicine platform.
Collapse
Affiliation(s)
- Won-Suk Lee
- From the Department of Surgery, Gil Medical Center, Gachon University (WSL, YHP, SYK); Lee Gil Ya Cancer and Diabetes Institute, Gachon University (WSL, HYK, HHJ, SYK, SH); Department of Hemato-Oncology, Gil Medical Center, Gachon University (DBS); Department of Pathology, Gil Medical Center, Gachon University (JYS); and Gachon Medical Research Institute, Gil Medical Center, Incheon, Korea (HHJ, SH)
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Epothilones A and B are naturally occurring microtubule stabilizers with nanomolar or even sub-nanomolar activity against human cancer cells in vitro and potent in vivo antitumor activity against multidrug-resistant tumors. Over the last decade, ten epothilonetype agents have entered clinical trials in humans; of these, the epothilone B lactam ixabepilone (BMS-247550; Ixempra®) was approved by the FDA for breast cancer treatment in 2007. Numerous synthetic and semisynthetic analogs of epothilones have been prepared and their in vitro and (in selected cases) in vivo biological activity has been determined, producing a wealth of SAR information on this compound family. This chapter will provide a brief summary of the in vitro and in vivo biological properties of epothilone B (Epo B). The major part of the discussion will then be organized around those epothilone analogs that have entered clinical development. For each analog the underlying synthetic chemistry and the most important preclinical features will be reviewed, together with the properties of some important related structures.
Collapse
Affiliation(s)
- Raphael Schiess
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich HCI H405, Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich HCI H405, Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| |
Collapse
|
15
|
Rosfjord E, Lucas J, Li G, Gerber HP. Advances in patient-derived tumor xenografts: from target identification to predicting clinical response rates in oncology. Biochem Pharmacol 2014; 91:135-43. [PMID: 24950467 DOI: 10.1016/j.bcp.2014.06.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 02/08/2023]
Abstract
Most oncology compounds entering clinical development have passed stringent preclinical pharmacology evaluation criteria. However, only a small fraction of experimental agents induce meaningful antitumor activities in the clinic. Low predictability of conventional preclinical pharmacology models is frequently cited as a main reason for the unusually high clinical attrition rates of therapeutic compounds in oncology. Therefore, improvement in the predictive values of preclinical efficacy models for clinical outcome holds great promise to reduce the clinical attrition rates of experimental compounds. Recent reports suggest that pharmacology studies conducted with patient derived xenograft (PDX) tumors are more predictive for clinical outcome compared to conventional, cell line derived xenograft (CDX) models, in particular when therapeutic compounds were tested at clinically relevant doses (CRDs). Moreover, the study of the most malignant cell types within tumors, the tumor initiating cells (TICs), relies on the availability of preclinical models that mimic the lineage hierarchy of cells within tumors. PDX models were shown to more closely recapitulate the heterogeneity of patient tumors and maintain the molecular, genetic, and histological complexity of human tumors during early stages of sequential passaging in mice, rendering them ideal tools to study the responses of TICs, tumor- and stromal cells to therapeutic intervention. In this commentary, we review the progress made in the development of PDX models in key areas of oncology research, including target identification and validation, tumor indication search and the development of a biomarker hypothesis that can be tested in the clinic to identify patients that will benefit most from therapeutic intervention.
Collapse
Affiliation(s)
- Edward Rosfjord
- Bioconjugate Discovery and Development, Oncology Research Units, 401 North Middletown Road, Pearl River, NY 10965, United States; Pfizer Worldwide Research and Development, United States
| | - Judy Lucas
- Bioconjugate Discovery and Development, Oncology Research Units, 401 North Middletown Road, Pearl River, NY 10965, United States; Pfizer Worldwide Research and Development, United States
| | - Gang Li
- Bioconjugate Discovery and Development, Oncology Research Units, 401 North Middletown Road, Pearl River, NY 10965, United States; Pfizer Worldwide Research and Development, United States
| | - Hans-Peter Gerber
- Bioconjugate Discovery and Development, Oncology Research Units, 401 North Middletown Road, Pearl River, NY 10965, United States; Pfizer Worldwide Research and Development, United States.
| |
Collapse
|
16
|
Rohena CC, Mooberry SL. Recent progress with microtubule stabilizers: new compounds, binding modes and cellular activities. Nat Prod Rep 2014; 31:335-55. [PMID: 24481420 PMCID: PMC4167679 DOI: 10.1039/c3np70092e] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nature has yielded numerous classes of chemically distinct microtubule stabilizers. Several of these, including paclitaxel (Taxol) and docetaxel (Taxotere), are important drugs used in the treatment of cancer. New microtubule stabilizers and novel formulations of these agents continue to provide advances in cancer therapy. In this review we cover recent progress in the chemistry and biology of these diverse microtubule stabilizers focusing on the wide range of organisms that produce these compounds, their mechanisms of inhibiting microtubule-dependent processes, mechanisms of drug resistance, and their interactions with tubulin including their distinct binding sites and modes. A new potential role for microtubule stabilizers in neurodegenerative diseases is reviewed.
Collapse
Affiliation(s)
- Cristina C. Rohena
- University of Texas Health Science Center at San Antonio,
7703 Floyd Curl Dr, San Antonio, TX, USA. Fax: 1(210)567-4300; Tel: 1(210) 567-6674;
| | - Susan L. Mooberry
- University of Texas Health Science Center at San Antonio,
7703 Floyd Curl Dr, San Antonio, TX, USA. Fax: 1(210)567-4300; Tel: 1(210) 567-6674;
- Cancer Therapy Research Center, 7979 Wurzbach Rd, San
Antonio, TX USA. Fax: 1(210)567-4300; Tel: 1(210) 567-4788;
| |
Collapse
|
17
|
Rogalska A, Gajek A, Marczak A. Analysis of epothilone B-induced cell death in normal ovarian cells. Cell Biol Int 2013; 37:1330-9. [DOI: 10.1002/cbin.10165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 07/22/2013] [Indexed: 01/15/2023]
Affiliation(s)
- Aneta Rogalska
- Department of Thermobiology; Institute of Biophysics; Faculty of Biology and Environmental Protection; University of Lodz; Pomorska 141/143 90-236 Lodz Poland
| | - Arkadiusz Gajek
- Department of Thermobiology; Institute of Biophysics; Faculty of Biology and Environmental Protection; University of Lodz; Pomorska 141/143 90-236 Lodz Poland
| | - Agnieszka Marczak
- Department of Thermobiology; Institute of Biophysics; Faculty of Biology and Environmental Protection; University of Lodz; Pomorska 141/143 90-236 Lodz Poland
| |
Collapse
|
18
|
Rogalska A, Szula E, Gajek A, Marczak A, Jóźwiak Z. Activation of apoptotic pathway in normal, cancer ovarian cells by epothilone B. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2013; 36:600-610. [PMID: 23838010 DOI: 10.1016/j.etap.2013.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/10/2013] [Accepted: 06/11/2013] [Indexed: 06/02/2023]
Abstract
The epothilones, a new class of microtubule-targeting agents, seem to be a very promising alternative to the current strategy of cancer treatment. We have analyzed the aspects of epothilone B (Epo B) on cellular metabolism of tumor (OV-90) and normal (MM 14) ovarian cells. The observed effects were compared with those of paclitaxel (PTX), which is now a standard for the treatment of ovarian cancer. The results provide direct evidence that Epo B is considerably more cytotoxic to human OV-90 ovarian cancer cells than PTX. We have found, that antitumor efficacy of this new drug is related to its apoptosis-inducing ability, which was confirmed during measurements typical markers of the process. Epo B induced changes in morphology of cells, mitochondrial membrane potential and cytochrome c release. Also a slight increase of the intracellular calcium level was observed. Moreover, we have found that ROS production, stimulated by Epo B, is directly involved in the induction of apoptosis via mitochondrial pathway.
Collapse
Affiliation(s)
- Aneta Rogalska
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Ewa Szula
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Arkadiusz Gajek
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Agnieszka Marczak
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Zofia Jóźwiak
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
19
|
Zhang XC, Zhang J, Li M, Huang XS, Yang XN, Zhong WZ, Xie L, Zhang L, Zhou M, Gavine P, Su X, Zheng L, Zhu G, Zhan P, Ji Q, Wu YL. Establishment of patient-derived non-small cell lung cancer xenograft models with genetic aberrations within EGFR, KRAS and FGFR1: useful tools for preclinical studies of targeted therapies. J Transl Med 2013; 11:168. [PMID: 23842453 PMCID: PMC3716998 DOI: 10.1186/1479-5876-11-168] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 07/08/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Patient-derived tumor xenograft models have been established and increasingly used for preclinical studies of targeted therapies in recent years. However, patient-derived non-small cell lung cancer (NSCLC) xenograft mouse models are relatively few in number and are limited in their degree of genetic characterization and validation. In this study, we aimed to establish a variety of patient-derived NSCLC models and characterize these for common genetic aberrations to provide more informative models for preclinical drug efficacy testing. METHODS NSCLC tissues from thirty-one patients were collected and implanted into immunodeficient mice. Established xenograft models were characterized for common genetic aberrations, including detection of gene mutations within EGFR and KRAS, and genetic amplification of FGFR1 and cMET. Finally, gefitinib anti-tumor efficacy was tested in these patient-derived NSCLC xenograft models. RESULTS Ten passable patient-derived NSCLC xenograft models were established by implantation of NSCLC specimens of thirty-one patients into immunodeficient mice. Genetic aberrations were detected in six of the models, including one model with an EGFR activating mutation (Exon19 Del), one model with KRAS mutation, one model with both KRAS mutation and cMET gene amplification, and three models with FGFR1 amplification. Anti-tumor efficacy studies using gefitinib demonstrated that the EGFR activating mutation model had superior sensitivity and that the KRAS mutation models were resistant to gefitinib. The range of gefitinib responses in the patient-derived NSCLC xenograft models were consistent with the results reported from clinical trials. Furthermore, we observed that patient-derived NSCLC models with FGFR1 gene amplification were insensitive to gefitinib treatment. CONCLUSIONS Ten patient-derived NSCLC xenograft models were established containing a variety of genetic aberrations including EGFR activating mutation, KRAS mutation, and FGFR1 and cMET amplification. Gefitinib anti-tumor efficacy in these patient-derived NSCLC xenografts containing EGFR and KRAS mutation was consistent with the reported results from previous clinical trials. Thus, data from our panel of patient-derived NSCLC xenograft models confirms the utility of these models in furthering our understanding of this disease and aiding the development of personalized therapies for NSCLC patients.
Collapse
Affiliation(s)
- Xu-chao Zhang
- Medical Research Center of Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangdong Lung Cancer Institute Guangzhou 510080, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Progress in oncology drug development has been hampered by a lack of preclinical models that reliably predict clinical activity of novel compounds in cancer patients. In an effort to address these shortcomings, there has been a recent increase in the use of patient-derived tumour xenografts (PDTX) engrafted into immune-compromised rodents such as athymic nude or NOD/SCID mice for preclinical modelling. Numerous tumour-specific PDTX models have been established and, importantly, they are biologically stable when passaged in mice in terms of global gene-expression patterns, mutational status, metastatic potential, drug responsiveness and tumour architecture. These characteristics might provide significant improvements over standard cell-line xenograft models. This Review will discuss specific PDTX disease examples illustrating an overview of the opportunities and limitations of these models in cancer drug development, and describe concepts regarding predictive biomarker development and future applications.
Collapse
|
21
|
Patient-derived xenografts of non small cell lung cancer: resurgence of an old model for investigation of modern concepts of tailored therapy and cancer stem cells. J Biomed Biotechnol 2012; 2012:568567. [PMID: 22547927 PMCID: PMC3324927 DOI: 10.1155/2012/568567] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 01/10/2012] [Indexed: 12/13/2022] Open
Abstract
Current chemotherapy regimens have unsatisfactory results in most advanced solid tumors. It is therefore imperative to devise novel therapeutic strategies and to optimize selection of patients, identifying early those who could benefit from available treatments. Mouse models are the most valuable tool for preclinical evaluation of novel therapeutic strategies in cancer and, among them, patient-derived xenografts models (PDX) have made a recent comeback in popularity. These models, obtained by direct implants of tissue fragments in immunocompromised mice, have great potential in drug development studies because they faithfully reproduce the patient's original tumor for both immunohistochemical markers and genetic alterations as well as in terms of response to common therapeutics They also maintain the original tumor heterogeneity, allowing studies of specific cellular subpopulations, including their modulation after drug treatment. Moreover PDXs maintain at least some aspects of the human microenvironment for weeks with the complete substitution with murine stroma occurring only after 2-3 passages in mouse and represent therefore a promising model for studies of tumor-microenvironment interaction. This review summarizes our present knowledge on mouse preclinical cancer models, with a particular attention on patient-derived xenografts of non small cell lung cancer and their relevance for preclinical and biological studies.
Collapse
|
22
|
Kapp FG, Sommer A, Kiefer T, Dölken G, Haendler B. 5-alpha-reductase type I (SRD5A1) is up-regulated in non-small cell lung cancer but does not impact proliferation, cell cycle distribution or apoptosis. Cancer Cell Int 2012; 12:1. [PMID: 22257483 PMCID: PMC3269976 DOI: 10.1186/1475-2867-12-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 01/18/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is one of the most frequent malignancies and has a high mortality rate due to late detection and lack of efficient treatments. Identifying novel drug targets for this indication may open the way for new treatment strategies. Comparison of gene expression profiles of NSCLC and normal adjacent tissue (NAT) allowed to determine that 5-alpha-reductase type I (SRD5A1) was up-regulated in NSCLC compared to NAT. This raised the question whether SRD5A1 was involved in sustained proliferation and survival of NSCLC. METHODS siRNA-mediated silencing of SRD5A1 was performed in A549 and NCI-H460 lung cancer cell lines in order to determine the impact on proliferation, on distribution during the different phases of the cell cycle, and on apoptosis/necrosis. In addition, lung cancer cell lines were treated with 4-azasteroids, which specifically inhibit SRD5A1 activity, and the effects on proliferation were measured. Statistical analyses using ANOVA and post-hoc Tamhane-T2-test were performed. In the case of non-parametric data, the Kruskal-Wallis test and the post-hoc Mann-Whitney-U-test were used. RESULTS The knock-down of SRDA51 expression was very efficient with the SRD5A1 transcripts being reduced to 10% of control levels. Knock-down efficiency was furthermore confirmed at the protein level. However, no effect of SRD5A1 silencing was observed in the proliferation assay, the cell cycle analysis, and the apoptosis/necrosis assay. Treatment of lung cancer cell lines with 4-azasteroids did not significantly inhibit proliferation. CONCLUSIONS In summary, the results suggest that SRD5A1 is not a crucial enzyme for the sustained proliferation of NSCLC cell lines.
Collapse
Affiliation(s)
- Friedrich G Kapp
- Global Drug Discovery, Bayer HealthCare, Müllerstr, 178, 13342 Berlin, Germany.
| | | | | | | | | |
Collapse
|
23
|
Petrul HM, Schatz CA, Kopitz CC, Adnane L, McCabe TJ, Trail P, Ha S, Chang YS, Voznesensky A, Ranges G, Tamburini PP. Therapeutic Mechanism and Efficacy of the Antibody–Drug Conjugate BAY 79-4620 Targeting Human Carbonic Anhydrase 9. Mol Cancer Ther 2011; 11:340-9. [DOI: 10.1158/1535-7163.mct-11-0523] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Araki K, Kitagawa K, Mukai H, Mukohara T, Kodama K, Ando Y, Narabayashi M, Minami H, Mera K, Sasaki Y. First clinical pharmacokinetic dose-escalation study of sagopilone, a novel, fully synthetic epothilone, in Japanese patients with refractory solid tumors. Invest New Drugs 2011; 30:2327-33. [DOI: 10.1007/s10637-011-9773-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 11/23/2011] [Indexed: 10/14/2022]
|
25
|
Eschenbrenner J, Winsel S, Hammer S, Sommer A, Mittelstaedt K, Drosch M, Klar U, Sachse C, Hannus M, Seidel M, Weiss B, Merz C, Siemeister G, Hoffmann J. Evaluation of activity and combination strategies with the microtubule-targeting drug sagopilone in breast cancer cell lines. Front Oncol 2011; 1:44. [PMID: 22649765 PMCID: PMC3355879 DOI: 10.3389/fonc.2011.00044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/31/2011] [Indexed: 12/14/2022] Open
Abstract
Sagopilone, a fully synthetic epothilone, is a microtubule-stabilizing agent optimized for high in vitro and in vivo activity against a broad range of tumor models, including those resistant to paclitaxel and other systemic treatments. Sagopilone development is accompanied by translational research studies to evaluate the molecular mode of action, to recognize mechanisms leading to resistance, to identify predictive response biomarkers, and to establish a rationale for combination with different therapies. Here, we profiled sagopilone activity in breast cancer cell lines. To analyze the mechanisms of mitotic arrest and apoptosis and to identify additional targets and biomarkers, an siRNA-based RNAi drug modifier screen interrogating 300 genes was performed in four cancer cell lines. Defects of the spindle assembly checkpoint (SAC) were identified to cause resistance against sagopilone-induced mitotic arrest and apoptosis. Potential biomarkers for resistance could therefore be functional defects like polymorphisms or mutations in the SAC, particularly in the central SAC kinase BUB1B. Moreover, chromosomal heterogeneity and polyploidy are also potential biomarkers of sagopilone resistance since they imply an increased tolerance for aberrant mitosis. RNAi screening further demonstrated that the sagopilone-induced mitotic arrest can be enhanced by concomitant inhibition of mitotic kinesins, thus suggesting a potential combination therapy of sagopilone with a KIF2C (MCAK) kinesin inhibitor. However, the combination of sagopilone and inhibition of the prophase kinesin KIF11 (EG5) is antagonistic, indicating that the kinesin inhibitor has to be highly specific to bring about the required therapeutic benefit.
Collapse
Affiliation(s)
- Julia Eschenbrenner
- Global Drug Discovery, Therapeutic Research Group Oncology, Bayer Healthcare Pharmaceuticals Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Strube A, Suominen MI, Rissanen JP, Mumberg D, Klar U, Halleen JM, Käkönen SM. The anti-tumor agent sagopilone shows antiresorptive effects both in vitro and in vivo. Osteoporos Int 2011; 22:2887-93. [PMID: 21104229 DOI: 10.1007/s00198-010-1486-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 11/04/2010] [Indexed: 11/24/2022]
Abstract
UNLABELLED Sagopilone, a fully synthetic epothilone and very potent anti-tumor agent, has proved to be efficient in inhibiting bone destruction and tumor burden in a mouse model of breast cancer bone metastasis. In addition to its antiproliferative effects, this study shows direct effects of sagopilone on bone resorption and osteoclast activity. INTRODUCTION Sagopilone, a novel fully synthetic third-generation epothilone, has proved to be efficient in inhibiting bone destruction and tumor burden in a mouse model of breast cancer bone metastasis. The aim of this study was to investigate whether the effect was primarily due to sagopilone's antiproliferative effect and consequent inhibition of tumor cell growth, or if sagopilone exerts direct effects on bone resorption and osteoclast activity. METHODS Sagopilone was studied and compared to paclitaxel in vitro in human osteoclast differentiation and activity cultures. For studying the potential of sagopilone for inhibiting bone resorption in vivo, a mouse model of ovariectomy (ovx)-induced osteoporosis was utilized. RESULTS Sagopilone inhibited osteoclast differentiation and activity more efficiently than paclitaxel and showed less cytotoxicity. Whereas sagopilone showed inhibitory effects on human osteoclast differentiation and activity already at 5 and 15 nM, respectively, paclitaxel started to show effects only at 20 and 100 nM concentrations, respectively. Sagopilone treatment increased BMD In the mouse ovx model even though a non-optimized dose was used which is effective in tumor-bearing mice. CONCLUSION This is the first study to evaluate sagopilone's effects on bone resorption in non-cancerous situation. The evidence that sagopilone is beneficial for bone will strengthen the status of sagopilone as an anti-cancer compound compared to other microtubule stabilizing agents.
Collapse
Affiliation(s)
- A Strube
- Global Drug Discovery, Bayer Schering Pharma AG, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Winsel S, Sommer A, Eschenbrenner J, Mittelstaedt K, Klar U, Hammer S, Hoffmann J. Molecular mode of action and role of TP53 in the sensitivity to the novel epothilone sagopilone (ZK-EPO) in A549 non-small cell lung cancer cells. PLoS One 2011; 6:e19273. [PMID: 21559393 PMCID: PMC3084814 DOI: 10.1371/journal.pone.0019273] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 03/31/2011] [Indexed: 01/01/2023] Open
Abstract
Sagopilone, an optimized fully synthetic epothilone, is a microtubule-stabilizing
compound that has shown high in vitro and in
vivo activity against a broad range of human tumor models. We
analyzed the differential mechanism of action of sagopilone in non-small cell
lung cancer cell lines in vitro. Sagopilone inhibited
proliferation of non-small cell lung cancer cell lines at lower nanomolar
concentration. The treatment with sagopilone caused strong disturbances of
cellular cytoskeletal organization. Two concentration-dependent phenotypes were
observed. At 2.5 nM sagopilone or 4 nM paclitaxel an aneuploid phenotype occur
whereas a mitotic arrest phenotype was induced by 40 nM sagopilone or
paclitaxel. Interestingly, treatment with 2.5 nM of sagopilone effectively
inhibited cell proliferation, but - compared to high concentrations (40 nM) -
only marginally induced apoptosis. Treatment with a high versus a low
concentration of sagopilone or paclitaxel regulates a non-overlapping set of
genes, indicating that both phenotypes substantially differ from each other.
Genes involved in G2/M phase transition and the spindle assembly checkpoint,
like Cyclin B1 and BUBR1 were upregulated by treatment with 40 nM sagopilone.
Unexpectedly, also genes involved in DNA damage response were upregulated under
that treatment. In contrast, treatment of A549 cells with a low concentration of
sagopilone revealed an upregulation of direct transcriptional target genes of
TP53, like CDKN1A, MDM2, GADD45A, FAS. Knockdown of TP53, which inhibited the
transcriptional induction of TP53 target genes, led to a significant increase in
apoptosis induction in A549 cells when treated with a low concentration of
sagopilone. The results indicate that activation of TP53 and its downstream
effectors like CDKN1A by low concentrations of sagopilone is responsible for the
relative apoptosis resistance of A549 cells and might represent a mechanism of
resistance to sagopilone.
Collapse
Affiliation(s)
- Sebastian Winsel
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
- Institute for Chemistry-Biochemistry, Freie
Universität Berlin, Berlin, Germany
- Medical Biotechnology, VTT Technical Research
Centre of Finland, Turku, Finland
| | - Anette Sommer
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
| | - Julia Eschenbrenner
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
- Institut für Biotechnologie, Technische
Universität Berlin, Berlin, Germany
| | - Kevin Mittelstaedt
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
- Department of Medicine, The University of
Melbourne, Melbourne, Australia
| | - Ulrich Klar
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
| | - Stefanie Hammer
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
- * E-mail: (SH); (JH)
| | - Jens Hoffmann
- Global Drug Discovery, Bayer Healthcare,
Berlin, Germany
- Experimental Pharmacology,
Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- * E-mail: (SH); (JH)
| |
Collapse
|
28
|
Merk J, Rolff J, Dorn C, Leschber G, Fichtner I. Chemoresistance in non-small-cell lung cancer: can multidrug resistance markers predict the response of xenograft lung cancer models to chemotherapy? Eur J Cardiothorac Surg 2011; 40:e29-33. [PMID: 21420313 DOI: 10.1016/j.ejcts.2011.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 02/02/2011] [Accepted: 02/04/2011] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE In chemotherapy for non-small-cell lung cancer (NSCLC), some patients seem to exhibit an intrinsic resistance or develop an acquired resistance under treatment. Results on resistance markers for possible treatment failure as shown in studies on selected lung cancer cell lines could not be completely confirmed in clinical trials. As these conflicting data require further research, we created a model between cell culture and the clinical need to study this problem. METHODS Our study was based on patient-derived NSCLC xenografts in a mouse model, which revealed a high coincidence with the original tumour. Protein and messenger RNA (mRNA) expression of known resistance markers (breast cancer resistance protein (BCRP), multidrug resistance P-glycoprotein (MDR), lung cancer-related protein (LRP) and multidrug resistance protein 1 (MRP1)) were analysed by real-time polymerase chain reaction (PCR) and immunoblotting in 24 xenografts. Chemosensitivity to etoposide, carboplatin, gemcitabine, paclitaxel, cetuximab and erlotinib was determined in in vivo xenograft experiments and compared with the protein and mRNA expression of the multidrug resistance markers. RESULTS With the exception of a single correlation between chemosensitivity and mRNA expression of etoposide and bcrp (mRNA expression of BCRP), we found no significant correlation between the response rates and protein- and mRNA expression levels in our 24 xenografts. The present results indicate that in vivo expression levels of multidrug resistance proteins and their mRNAs may not play a comparable role in chemoresistance of NSCLC, as pointed out in selected tumour cell lines. CONCLUSIONS Patient-derived xenografts allow detailed investigation of therapy-related markers and their dynamic regulation in a well-standardised and clinically related way. As a consequence of our investigations, we regard multidrug resistance to be a multifactorial phenomenon, in which more factors than the markers analysed by the present study may be involved.
Collapse
Affiliation(s)
- Johannes Merk
- Department of Thoracic Surgery, ELK Berlin Chest Hospital, Berlin, Germany.
| | | | | | | | | |
Collapse
|
29
|
Langer W, Sohler F, Leder G, Beckmann G, Seidel H, Gröne J, Hummel M, Sommer A. Exon array analysis using re-defined probe sets results in reliable identification of alternatively spliced genes in non-small cell lung cancer. BMC Genomics 2010; 11:676. [PMID: 21118496 PMCID: PMC3053589 DOI: 10.1186/1471-2164-11-676] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 11/30/2010] [Indexed: 12/22/2022] Open
Abstract
Background Treatment of non-small cell lung cancer with novel targeted therapies is a major unmet clinical need. Alternative splicing is a mechanism which generates diverse protein products and is of functional relevance in cancer. Results In this study, a genome-wide analysis of the alteration of splicing patterns between lung cancer and normal lung tissue was performed. We generated an exon array data set derived from matched pairs of lung cancer and normal lung tissue including both the adenocarcinoma and the squamous cell carcinoma subtypes. An enhanced workflow was developed to reliably detect differential splicing in an exon array data set. In total, 330 genes were found to be differentially spliced in non-small cell lung cancer compared to normal lung tissue. Microarray findings were validated with independent laboratory methods for CLSTN1, FN1, KIAA1217, MYO18A, NCOR2, NUMB, SLK, SYNE2, TPM1, (in total, 10 events) and ADD3, which was analysed in depth. We achieved a high validation rate of 69%. Evidence was found that the activity of FOX2, the splicing factor shown to cause cancer-specific splicing patterns in breast and ovarian cancer, is not altered at the transcript level in several cancer types including lung cancer. Conclusions This study demonstrates how alternatively spliced genes can reliably be identified in a cancer data set. Our findings underline that key processes of cancer progression in NSCLC are affected by alternative splicing, which can be exploited in the search for novel targeted therapies.
Collapse
Affiliation(s)
- Wolfram Langer
- Bayer Schering Pharma AG, Global Drug Discovery (GDD)-Target Discovery, Müllerstrasse 178, 13342 Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin Transl Oncol 2010; 12:473-80. [PMID: 20615824 DOI: 10.1007/s12094-010-0540-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mouse cancer models have consistently been used to qualify new anticancer drugs in the development of human clinical trials. Rodent tumour models currently being used and which include transgenic tumour models, and those generated by planting human tumour cell lines subcutaneously in immunodeficient mice, do not sufficiently represent clinical cancer characteristics, especially with regard to metastasis and drug sensitivity. The increasingly used patient-derived human tumour tissue (PDTT) xenografts models implanted subcutaneously or in subrenal capsule in immunodeficient mice, such as athymic nude mice or severe combined immunedeficient (SCID) mice, may provide a more accurate reflection of human tumour biological characteristics than tumour cell lines. The ability to passage patients' fresh tumour tissues into large numbers of immunodeficient mice provides possibilities for better preclinical testing of new therapies for the treatment and better outcome for cancer. In this review, we outline the methods of establishing xenograft models, discuss the biological stability of PDTT xenograft models and demonstrate their roles in developing new anticancer drugs and testing therapeutic regimens in cancer patients.
Collapse
Affiliation(s)
- Ketao Jin
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | |
Collapse
|
31
|
Jin K, He K, Li G, Teng L. Personalized cancer therapy using a patient-derived tumor tissue xenograft model: a translational field worthy of exploring further? Per Med 2010; 7:597-606. [PMID: 29776245 DOI: 10.2217/pme.10.48] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It has long been observed that interpatient variability in response to anticancer drugs is associated with different outcomes. Oncologists continually hold the desire of matching the right therapeutic regimen with the right cancer patient, which is termed ‘personalized cancer therapy’. Rapid advances in genetics, genomics and related technologies are promising a new era of personalized cancer therapy based on individual molecular biomarkers. However, these molecular predictors of tumor response are far from perfect. Because of the inherent limitations in the current approaches for anticancer drugs response prediction, the need for new techniques to predict tumor response to therapy is urgent. Using a patient-derived human tumor tissue (PDTT) xenograft model to predict tumor response to therapy might be an ideal candidate method to choose. This article provides an overview of the achievements and limitations of genetic, genomic and proteomic molecular markers for personalized cancer therapy, and further discusses the potentials of using a PDTT xenograft model as a candidate strategy for personalized cancer therapy.
Collapse
Affiliation(s)
- Ketao Jin
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of General Surgery, Zhejiang University Teaching Hospital Zhuji Hospital, Zhuji, Zhejiang, China
| | - Kuifeng He
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangliang Li
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | |
Collapse
|