451
|
Pazopanib, a Receptor Tyrosine Kinase Inhibitor, Suppresses Tumor Growth through Angiogenesis in Dedifferentiated Liposarcoma Xenograft Models. Transl Oncol 2014; 7:665-71. [PMID: 25500074 PMCID: PMC4311036 DOI: 10.1016/j.tranon.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/15/2014] [Accepted: 09/19/2014] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION The rarity of dedifferentiated liposarcoma (DDLPS) and the lack of experimental DDLPS models limit the development of novel therapeutic strategies. Pazopanib (PAZ) is a tyrosine kinase inhibitor that is approved for the treatment of non-adipocytic advanced soft tissue sarcoma. The activity of this agent has not yet been properly explored in preclinical liposarcoma models nor in a randomized phase Ш clinical trial in this entity. The aim of the present study was to investigate whether PAZ had antitumor activity in DDLPS models in vivo. MATERIAL AND METHODS We established two patient-derived DDLPS xenograft models (UZLX-STS3 and UZLX-STS5) through implantation of tumor material from sarcoma patients in athymic nude NMRI mice. An animal model of the SW872 liposarcoma cell line was also used. To investigate the efficacy of PAZ in vivo, mice bearing tumors were treated for 2 weeks with sterile water, doxorubicin (1.2 mg/kg, intraperitoneally, twice per week), PAZ [40 mg/kg, orally (p.o.), twice per day], or PAZ plus doxorubicin (same schedules as for single treatments). RESULTS Patient-derived xenografts retained the histologic and molecular features of DDLPS. PAZ significantly delayed tumor growth by decreasing proliferation and inhibited angiogenesis in all models tested. Combining the angiogenesis inhibitor with an anthracycline did not show superior efficacy. CONCLUSION These results suggest that PAZ has potential antitumor activity in DDLPS primarily through antiangiogenic effects and therefore should be explored in clinical trials.
Collapse
|
452
|
3D tissue-engineered model of Ewing's sarcoma. Adv Drug Deliv Rev 2014; 79-80:155-71. [PMID: 25109853 DOI: 10.1016/j.addr.2014.07.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/28/2014] [Accepted: 07/24/2014] [Indexed: 12/30/2022]
Abstract
Despite longstanding reliance upon monolayer culture for studying cancer cells, and numerous advantages from both a practical and experimental standpoint, a growing body of evidence suggests that more complex three-dimensional (3D) models are necessary to properly mimic many of the critical hallmarks associated with the oncogenesis, maintenance and spread of Ewing's sarcoma (ES), the second most common pediatric bone tumor. And as clinicians increasingly turn to biologically-targeted therapies that exert their effects not only on the tumor cells themselves, but also on the surrounding extracellular matrix, it is especially important that preclinical models evolve in parallel to reliably measure antineoplastic effects and possible mechanisms of de novo and acquired drug resistance. Herein, we highlight a number of innovative methods used to fabricate biomimetic ES tumors, encompassing both the surrounding cellular milieu and the extracellular matrix (ECM), and suggest potential applications to advance our understanding of ES biology, preclinical drug testing, and personalized medicine.
Collapse
|
453
|
Guilhamon P, Butcher LM, Presneau N, Wilson GA, Feber A, Paul DS, Schütte M, Haybaeck J, Keilholz U, Hoffman J, Ross MT, Flanagan AM, Beck S. Assessment of patient-derived tumour xenografts (PDXs) as a discovery tool for cancer epigenomics. Genome Med 2014; 6:116. [PMID: 25587359 PMCID: PMC4292812 DOI: 10.1186/s13073-014-0116-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The use of tumour xenografts is a well-established research tool in cancer genomics but has not yet been comprehensively evaluated for cancer epigenomics. METHODS In this study, we assessed the suitability of patient-derived tumour xenografts (PDXs) for methylome analysis using Infinium 450 K Beadchips and MeDIP-seq. RESULTS Controlled for confounding host (mouse) sequences, comparison of primary PDXs and matching patient tumours in a rare (osteosarcoma) and common (colon) cancer revealed that an average 2.7% of the assayed CpG sites undergo major (Δβ ≥ 0.51) methylation changes in a cancer-specific manner as a result of the xenografting procedure. No significant subsequent methylation changes were observed after a second round of xenografting between primary and secondary PDXs. Based on computational simulation using publically available methylation data, we additionally show that future studies comparing two groups of PDXs should use 15 or more samples in each group to minimise the impact of xenografting-associated changes in methylation on comparison results. CONCLUSIONS Our results from rare and common cancers indicate that PDXs are a suitable discovery tool for cancer epigenomics and we provide guidance on how to overcome the observed limitations.
Collapse
Affiliation(s)
- Paul Guilhamon
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK
| | - Lee M Butcher
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK
| | - Nadege Presneau
- Genetics and Cell Biology of Sarcoma, UCL Cancer Institute, University College London, London, WC1E 6DD UK ; Department of Biomedical Sciences, University of Westminster, London, W1W 6UW UK
| | - Gareth A Wilson
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK ; Translational Cancer Therapeutics Laboratory, CR-UK London Research Institute, London, WC2A 3LY UK
| | - Andrew Feber
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK
| | - Dirk S Paul
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK
| | | | - Johannes Haybaeck
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Ulrich Keilholz
- Department of Hematology and Medical Oncology, Charité Comprehensive Cancer Center, D-10117 Berlin, Germany
| | | | - Mark T Ross
- Illumina Cambridge Ltd, Chesterford Research Park, Little Chesterford, CB10 1XL UK
| | - Adrienne M Flanagan
- Genetics and Cell Biology of Sarcoma, UCL Cancer Institute, University College London, London, WC1E 6DD UK ; Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, Middlesex, London, HA7 4LP UK
| | - Stephan Beck
- Medical Genomics, UCL Cancer Institute, University College London, London, WC1E 6DD UK
| |
Collapse
|
454
|
Nakajima A, Endo H, Okuyama H, Kiyohara Y, Kimura T, Kamiura S, Hiraoka M, Inoue M. Radiation sensitivity assay with a panel of patient-derived spheroids of small cell carcinoma of the cervix. Int J Cancer 2014; 136:2949-60. [PMID: 25408479 DOI: 10.1002/ijc.29349] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/11/2014] [Indexed: 11/11/2022]
Abstract
Small cell carcinoma of the uterine cervix (SCCC) is a rare cancer with a poor prognosis for which no standard treatment exists. Here, we successfully established panels of patient-derived spheroid cultures from six SCCC patient samples by cancer tissue-originated spheroids (CTOS) method. To assess the intrinsic radiosensitivity and mechanism of radioresistance in individual SCCC patients, we further developed an in vitro sensitivity assay for radiation. Radiation sensitivity in the CTOS assay varied among individual cases and was consistent with in vivo radiation sensitivity using CTOS-derived xenograft tumors in the examined cases. Furthermore, by comparing gene expression in CTOSs with different radiosensitivity, we found that expression of hypoxia-inducible factor-1α (HIF-1α) target genes was upregulated in resistant CTOSs. HIF-1α protein levels increased several hours after irradiation. In a radioresistant CTOS, an inhibitor of heat shock protein 90 (HSP90) suppressed radiation-induced HIF-1α expression. Suppression of HIF-1α by small hairpin RNA significantly enhanced the effect of radiation, at least in part by promoting radiation-induced apoptosis. HSP90 inhibitor also increased radiation sensitivity. Our results indicate that radiation-induced HIF-1α upregulation was one mechanism of radioresistance in a radioresistant SCCC CTOS. Accumulating CTOS lines may provide a good platform to study characters of rare cancers like SCCC.
Collapse
Affiliation(s)
- Aya Nakajima
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
455
|
Burgenske DM, Monsma DJ, Dylewski D, Scott SB, Sayfie AD, Kim DG, Luchtefeld M, Martin KR, Stephenson P, Hostetter G, Dujovny N, MacKeigan JP. Establishment of genetically diverse patient-derived xenografts of colorectal cancer. Am J Cancer Res 2014; 4:824-837. [PMID: 25520871 PMCID: PMC4266715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/20/2014] [Indexed: 06/04/2023] Open
Abstract
Preclinical compounds tested in animal models often show limited efficacy when transitioned into human clinical trials. As a result, many patients are stratified into treatment regimens that have little impact on their disease. In order to create preclinical models that can more accurately predict tumor responses, we established patient-derived xenograft (PDX) models of colorectal cancer (CRC). Surgically resected tumor specimens from colorectal cancer patients were implanted subcutaneously into athymic nude mice. Following successful establishment, fourteen models underwent further evaluation to determine whether these models exhibit heterogeneity, both at the cellular and genetic level. Histological review revealed properties not found in CRC cell lines, most notably in overall architecture (predominantly columnar epithelium with evidence of gland formation) and the presence of mucin-producing cells. Custom CRC gene panels identified somatic driver mutations in each model, and therapeutic efficacy studies in tumor-bearing mice were designed to determine how models with known mutations respond to PI3K, mTOR, or MAPK inhibitors. Interestingly, MAPK pathway inhibition drove tumor responses across most models tested. Noteworthy, the MAPK inhibitor PD0325901 alone did not significantly mediate tumor response in the context of a KRAS(G12D) model, and improved tumor responses resulted when combined with mTOR inhibition. As a result, these genetically diverse models represent a valuable resource for preclinical efficacy and drug discovery studies.
Collapse
Affiliation(s)
- Danielle M Burgenske
- Laboratory of Systems Biology, Van Andel Research InstituteGrand Rapids, MI 49503, USA
- Van Andel Institute Graduate SchoolGrand Rapids, MI 49503, USA
| | - David J Monsma
- Preclinical Therapeutics, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Dawna Dylewski
- Preclinical Therapeutics, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Stephanie B Scott
- Preclinical Therapeutics, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Aaron D Sayfie
- Laboratory of Systems Biology, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Donald G Kim
- Ferguson-Blodgett Digestive Disease Institute, Spectrum Health Medical GroupGrand Rapids, MI 49503, USA
| | - Martin Luchtefeld
- Ferguson-Blodgett Digestive Disease Institute, Spectrum Health Medical GroupGrand Rapids, MI 49503, USA
| | - Katie R Martin
- Laboratory of Systems Biology, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Paul Stephenson
- Department of Statistics, Grand Valley State UniversityAllendale, MI 49401, USA
| | - Galen Hostetter
- Laboratory of Analytical Pathology, Van Andel Research InstituteGrand Rapids, MI 49503, USA
| | - Nadav Dujovny
- Ferguson-Blodgett Digestive Disease Institute, Spectrum Health Medical GroupGrand Rapids, MI 49503, USA
| | - Jeffrey P MacKeigan
- Laboratory of Systems Biology, Van Andel Research InstituteGrand Rapids, MI 49503, USA
- Van Andel Institute Graduate SchoolGrand Rapids, MI 49503, USA
| |
Collapse
|
456
|
Pettersen EO, Ebbesen P, Gieling RG, Williams KJ, Dubois L, Lambin P, Ward C, Meehan J, Kunkler IH, Langdon SP, Ree AH, Flatmark K, Lyng H, Calzada MJ, Peso LD, Landazuri MO, Görlach A, Flamm H, Kieninger J, Urban G, Weltin A, Singleton DC, Haider S, Buffa FM, Harris AL, Scozzafava A, Supuran CT, Moser I, Jobst G, Busk M, Toustrup K, Overgaard J, Alsner J, Pouyssegur J, Chiche J, Mazure N, Marchiq I, Parks S, Ahmed A, Ashcroft M, Pastorekova S, Cao Y, Rouschop KM, Wouters BG, Koritzinsky M, Mujcic H, Cojocari D. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: the METOXIA consortium. J Enzyme Inhib Med Chem 2014; 30:689-721. [PMID: 25347767 DOI: 10.3109/14756366.2014.966704] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 01/06/2023] Open
Abstract
The hypoxic areas of solid cancers represent a negative prognostic factor irrespective of which treatment modality is chosen for the patient. Still, after almost 80 years of focus on the problems created by hypoxia in solid tumours, we still largely lack methods to deal efficiently with these treatment-resistant cells. The consequences of this lack may be serious for many patients: Not only is there a negative correlation between the hypoxic fraction in tumours and the outcome of radiotherapy as well as many types of chemotherapy, a correlation has been shown between the hypoxic fraction in tumours and cancer metastasis. Thus, on a fundamental basis the great variety of problems related to hypoxia in cancer treatment has to do with the broad range of functions oxygen (and lack of oxygen) have in cells and tissues. Therefore, activation-deactivation of oxygen-regulated cascades related to metabolism or external signalling are important areas for the identification of mechanisms as potential targets for hypoxia-specific treatment. Also the chemistry related to reactive oxygen radicals (ROS) and the biological handling of ROS are part of the problem complex. The problem is further complicated by the great variety in oxygen concentrations found in tissues. For tumour hypoxia to be used as a marker for individualisation of treatment there is a need for non-invasive methods to measure oxygen routinely in patient tumours. A large-scale collaborative EU-financed project 2009-2014 denoted METOXIA has studied all the mentioned aspects of hypoxia with the aim of selecting potential targets for new hypoxia-specific therapy and develop the first stage of tests for this therapy. A new non-invasive PET-imaging method based on the 2-nitroimidazole [(18)F]-HX4 was found to be promising in a clinical trial on NSCLC patients. New preclinical models for testing of the metastatic potential of cells were developed, both in vitro (2D as well as 3D models) and in mice (orthotopic grafting). Low density quantitative real-time polymerase chain reaction (qPCR)-based assays were developed measuring multiple hypoxia-responsive markers in parallel to identify tumour hypoxia-related patterns of gene expression. As possible targets for new therapy two main regulatory cascades were prioritised: The hypoxia-inducible-factor (HIF)-regulated cascades operating at moderate to weak hypoxia (<1% O(2)), and the unfolded protein response (UPR) activated by endoplasmatic reticulum (ER) stress and operating at more severe hypoxia (<0.2%). The prioritised targets were the HIF-regulated proteins carbonic anhydrase IX (CAIX), the lactate transporter MCT4 and the PERK/eIF2α/ATF4-arm of the UPR. The METOXIA project has developed patented compounds targeting CAIX with a preclinical documented effect. Since hypoxia-specific treatments alone are not curative they will have to be combined with traditional anti-cancer therapy to eradicate the aerobic cancer cell population as well.
Collapse
|
457
|
Using the Lessons Learned From the Clinic to Improve the Preclinical Development of Antibody Drug Conjugates. Pharm Res 2014; 32:3458-69. [PMID: 25339341 PMCID: PMC4596896 DOI: 10.1007/s11095-014-1536-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/29/2014] [Indexed: 12/22/2022]
Abstract
The treatment options for cancer patients include surgery, chemotherapeutics, radiation therapy, antibody therapy and various combinations of these therapies. The challenge with each therapy is finding the balance between maximizing the anti-tumor efficacy while minimizing the dose limiting toxicities. Antibodies, unlike small molecule chemotherapeutics, selectively bind to cell surface tumor antigens and can be used to deliver radionucleotides or small molecule chemotherapeutic drugs directly to the tumor. Advances in antibody engineering, linker chemistry and the identification of potent cytotoxic drugs led to the recent approval of two antibody drug conjugates to treat breast cancer and lymphoma patients. We will discuss how the observations from the clinical development of antibody drug conjugates can guide the preclinical development of the next generation of antibody drug conjugates.
Collapse
|
458
|
Bult CJ, Krupke DM, Begley DA, Richardson JE, Neuhauser SB, Sundberg JP, Eppig JT. Mouse Tumor Biology (MTB): a database of mouse models for human cancer. Nucleic Acids Res 2014; 43:D818-24. [PMID: 25332399 PMCID: PMC4384039 DOI: 10.1093/nar/gku987] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Mouse Tumor Biology (MTB; http://tumor.informatics.jax.org) database is a unique online compendium of mouse models for human cancer. MTB provides online access to expertly curated information on diverse mouse models for human cancer and interfaces for searching and visualizing data associated with these models. The information in MTB is designed to facilitate the selection of strains for cancer research and is a platform for mining data on tumor development and patterns of metastases. MTB curators acquire data through manual curation of peer-reviewed scientific literature and from direct submissions by researchers. Data in MTB are also obtained from other bioinformatics resources including PathBase, the Gene Expression Omnibus and ArrayExpress. Recent enhancements to MTB improve the association between mouse models and human genes commonly mutated in a variety of cancers as identified in large-scale cancer genomics studies, provide new interfaces for exploring regions of the mouse genome associated with cancer phenotypes and incorporate data and information related to Patient-Derived Xenograft models of human cancers.
Collapse
Affiliation(s)
- Carol J Bult
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Debra M Krupke
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Dale A Begley
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | | | - John P Sundberg
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Janan T Eppig
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| |
Collapse
|
459
|
Ricci F, Bizzaro F, Cesca M, Guffanti F, Ganzinelli M, Decio A, Ghilardi C, Perego P, Fruscio R, Buda A, Milani R, Ostano P, Chiorino G, Bani MR, Damia G, Giavazzi R. Patient-derived ovarian tumor xenografts recapitulate human clinicopathology and genetic alterations. Cancer Res 2014; 74:6980-90. [PMID: 25304260 DOI: 10.1158/0008-5472.can-14-0274] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy. On the basis of its histopathology and molecular-genomic changes, ovarian cancer has been divided into subtypes, each with distinct biology and outcome. The aim of this study was to develop a panel of patient-derived EOC xenografts that recapitulate the molecular and biologic heterogeneity of human ovarian cancer. Thirty-four EOC xenografts were successfully established, either subcutaneously or intraperitoneally, in nude mice. The xenografts were histologically similar to the corresponding patient tumor and comprised all the major ovarian cancer subtypes. After orthotopic transplantation in the bursa of the mouse ovary, they disseminate into the organs of the peritoneal cavity and produce ascites, typical of ovarian cancer. Gene expression analysis and mutation status indicated a high degree of similarity with the original patient and discriminate different subsets of xenografts. They were very responsive, responsive, and resistant to cisplatin, resembling the clinical situation in ovarian cancer. This panel of patient-derived EOC xenografts that recapitulate the recently type I and type II classification serves to study the biology of ovarian cancer, identify tumor-specific molecular markers, and develop novel treatment modalities.
Collapse
Affiliation(s)
- Francesca Ricci
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Francesca Bizzaro
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Marta Cesca
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Federica Guffanti
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Monica Ganzinelli
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Alessandra Decio
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Carmen Ghilardi
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | - Robert Fruscio
- Obstetrics and Gynecology Clinic, San Gerardo Hospital, Monza, Italy
| | - Alessandro Buda
- Obstetrics and Gynecology Clinic, San Gerardo Hospital, Monza, Italy
| | - Rodolfo Milani
- Obstetrics and Gynecology Clinic, San Gerardo Hospital, Monza, Italy
| | - Paola Ostano
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - Giovanna Chiorino
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - Maria Rosa Bani
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Giovanna Damia
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | - Raffaella Giavazzi
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| |
Collapse
|
460
|
Braekeveldt N, Wigerup C, Gisselsson D, Mohlin S, Merselius M, Beckman S, Jonson T, Börjesson A, Backman T, Tadeo I, Berbegall AP, Ora I, Navarro S, Noguera R, Påhlman S, Bexell D. Neuroblastoma patient-derived orthotopic xenografts retain metastatic patterns and geno- and phenotypes of patient tumours. Int J Cancer 2014; 136:E252-61. [PMID: 25220031 PMCID: PMC4299502 DOI: 10.1002/ijc.29217] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/31/2014] [Accepted: 09/01/2014] [Indexed: 12/24/2022]
Abstract
Neuroblastoma is a childhood tumour with heterogeneous characteristics and children with metastatic disease often have a poor outcome. Here we describe the establishment of neuroblastoma patient-derived xenografts (PDXs) by orthotopic implantation of viably cryopreserved or fresh tumour explants of patients with high risk neuroblastoma into immunodeficient mice. In vivo tumour growth was monitored by magnetic resonance imaging and fluorodeoxyglucose–positron emission tomography. Neuroblastoma PDXs retained the undifferentiated histology and proliferative capacity of their corresponding patient tumours. The PDXs expressed neuroblastoma markers neural cell adhesion molecule, chromogranin A, synaptophysin and tyrosine hydroxylase. Whole genome genotyping array analyses demonstrated that PDXs retained patient-specific chromosomal aberrations such as MYCN amplification, deletion of 1p and gain of chromosome 17q. Thus, neuroblastoma PDXs recapitulate the hallmarks of high-risk neuroblastoma in patients. PDX-derived cells were cultured in serum-free medium where they formed free-floating neurospheres, expressed neuroblastoma gene markers MYCN, CHGA, TH, SYP and NPY, and retained tumour-initiating and metastatic capacity in vivo. PDXs showed much higher degree of infiltrative growth and distant metastasis as compared to neuroblastoma SK-N-BE(2)c cell line-derived orthotopic tumours. Importantly, the PDXs presented with bone marrow involvement, a clinical feature of aggressive neuroblastoma. Thus, neuroblastoma PDXs serve as clinically relevant models for studying and targeting high-risk metastatic neuroblastoma.
Collapse
|
461
|
Doroshow JH, Kummar S. Translational research in oncology--10 years of progress and future prospects. Nat Rev Clin Oncol 2014; 11:649-62. [PMID: 25286976 DOI: 10.1038/nrclinonc.2014.158] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
International efforts to sequence the genomes of various human cancers have been broadly deployed in drug discovery programmes. Diagnostic tests that predict the value of the molecularly targeted anticancer agents used in such programmes are conceived and validated in parallel with new small-molecule treatments and immunotherapies. This approach has been aided by better preclinical cancer models; an enhanced appreciation of the complex interactions that exist between tumour cells and their microenvironment; the elucidation of interactions between many of the genetic drivers of cancer, including oncogenes and tumour suppressors; and recent insights into the genetic heterogeneity of human tumours made possible by extraordinary improvements in DNA-sequencing techniques. These advances are being employed in the first generation of genomic clinical trials that will examine the feasibility of matching a broad range of systemic therapies to specific molecular tumour characteristics. More-extensive molecular characterization of tumours and their supporting matrices are anticipated to become standard aspects of oncological practice, permitting continuous molecular re-evaluations of human malignancies on a patient-by-patient and treatment-by-treatment basis. We review selected developments in translational cancer biology, diagnostics, and therapeutics that have occurred over the past decade and offer our thoughts on future prospects for the next few years.
Collapse
Affiliation(s)
- James H Doroshow
- 1] Division of Cancer Treatment and Diagnosis, Room 3A-44, Building 31, 31 Center Drive, National Cancer Institute, NIH, Bethesda, MD 20892, USA. [2] Developmental Therapeutics Branch of the Center for Cancer Research, Room 3A-44, Building 31, 31 Center Drive, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, Room 3A-44, Building 31, 31 Center Drive, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
462
|
Bentley VL, Veinotte CJ, Corkery DP, Pinder JB, LeBlanc MA, Bedard K, Weng AP, Berman JN, Dellaire G. Focused chemical genomics using zebrafish xenotransplantation as a pre-clinical therapeutic platform for T-cell acute lymphoblastic leukemia. Haematologica 2014; 100:70-6. [PMID: 25281505 DOI: 10.3324/haematol.2014.110742] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer therapeutics is evolving to precision medicine, with the goal of matching targeted compounds with molecular aberrations underlying a patient's cancer. While murine models offer a pre-clinical tool, associated costs and time are not compatible with actionable patient-directed interventions. Using the paradigm of T-cell acute lymphoblastic leukemia, a high-risk disease with defined molecular underpinnings, we developed a zebrafish human cancer xenotransplantation model to inform therapeutic decisions. Using a focused chemical genomic approach, we demonstrate that xenografted cell lines harboring mutations in the NOTCH1 and PI3K/AKT pathways respond concordantly to their targeted therapies, patient-derived T-cell acute lymphoblastic leukemia can be successfully engrafted in zebrafish and specific drug responses can be quantitatively determined. Using this approach, we identified a mutation sensitive to γ-secretase inhibition in a xenograft from a child with T-cell acute lymphoblastic leukemia, confirmed by Sanger sequencing and validated as a gain-of-function NOTCH1 mutation. The zebrafish xenotransplantation platform provides a novel cost-effective means of tailoring leukemia therapy in real time.
Collapse
Affiliation(s)
| | | | - Dale P Corkery
- Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS
| | | | | | | | - Andrew P Weng
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC
| | - Jason N Berman
- IWK Health Centre, Halifax, NS Pediatrics and Microbiology & Immunology Dalhousie University, Halifax, NS, Canada
| | - Graham Dellaire
- Pathology, Dalhousie University, Halifax, NS Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS
| |
Collapse
|
463
|
Giannelli G, Rani B, Dituri F, Cao Y, Palasciano G. Moving towards personalised therapy in patients with hepatocellular carcinoma: the role of the microenvironment. Gut 2014; 63:1668-76. [PMID: 25053718 DOI: 10.1136/gutjnl-2014-307323] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The goal of personalised therapy based on hepatocellular carcinoma (HCC) molecular characteristics is still beyond our grasp. Systemic treatments show poor efficacy, mainly because of the great heterogeneity of the tumour. Indeed, differences in aetiology, disease stage and biochemical composition of the fibrotic liver make cirrhosis itself a highly dyshomogeneous disease. Cancer cells grow in a cirrhotic microenvironment, interacting with stromal cells and engaging matrix components that differ from patient to patient, hampering the development of drugs to treat all patients. Growing evidence suggests a role for the cross-talk between HCC and the host stroma in driving disease progression and hence prognosis and survival. Many efforts have been devoted to identifying genes responsible for good or bad prognosis, but no study has yet proven helpful in guiding therapeutic choices and management over time, also taking into account the development of drug resistance. The questions of what to target and in which patient are still unsolved. In the personalised therapy scenario, the patient rather than the disease becomes the target of the therapy. However, this still requires an evidence-based medical approach. Herein, we will discuss how individual differences in terms of quality and quantity of the tissue microenvironment components affect progression of HCC. Then, we will highlight potential druggable pathways, also considering ongoing clinical trials. The development of biomarkers will be discussed in the light of new experimental research conducted with the aim of moving towards personalised therapy in patients with HCC.
Collapse
Affiliation(s)
- Gianluigi Giannelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Bhavna Rani
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Francesco Dituri
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Yuan Cao
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Giuseppe Palasciano
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
464
|
Begley DA, Krupke DM, Neuhauser SB, Richardson JE, Schofield PN, Bult CJ, Eppig JT, Sundberg JP. Identifying mouse models for skin cancer using the Mouse Tumor Biology Database. Exp Dermatol 2014; 23:761-3. [PMID: 25040013 PMCID: PMC4183210 DOI: 10.1111/exd.12512] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2014] [Indexed: 11/29/2022]
Abstract
In recent years, the scientific community has generated an ever-increasing amount of data from a growing number of animal models of human cancers. Much of these data come from genetically engineered mouse models. Identifying appropriate models for skin cancer and related relevant genetic data sets from an expanding pool of widely disseminated data can be a daunting task. The Mouse Tumor Biology Database (MTB) provides an electronic archive, search and analysis system that can be used to identify dermatological mouse models of cancer, retrieve model-specific data and analyse these data. In this report, we detail MTB's contents and capabilities, together with instructions on how to use MTB to search for skin-related tumor models and associated data.
Collapse
Affiliation(s)
| | | | | | | | - Paul N. Schofield
- The Jackson Laboratory, Bar Harbor, ME USA
- Dept. of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
465
|
Goldenberg DM, Rooney RJ, Loo M, Liu D, Chang CH. In-vivo fusion of human cancer and hamster stromal cells permanently transduces and transcribes human DNA. PLoS One 2014; 9:e107927. [PMID: 25259521 PMCID: PMC4178054 DOI: 10.1371/journal.pone.0107927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/19/2014] [Indexed: 12/16/2022] Open
Abstract
After demonstrating, with karyotyping, polymerase chain reaction (PCR) and fluorescence in-situ hybridization, the retention of certain human chromosomes and genes following the spontaneous fusion of human tumor and hamster cells in-vivo, it was postulated that cell fusion causes the horizontal transmission of malignancy and donor genes. Here, we analyzed gene expression profiles of 3 different hybrid tumors first generated in the hamster cheek pouch after human tumor grafting, and then propagated in hamsters and in cell cultures for years: two Hodgkin lymphomas (GW-532, GW-584) and a glioblastoma multiforme (GB-749). Based on the criteria of MAS 5.0 detection P-values ≤0.065 and at least a 2-fold greater signal expression value than a hamster melanoma control, we identified 3,759 probe sets (ranging from 1,040 to 1,303 in each transplant) from formalin-fixed, paraffin-embedded sections of the 3 hybrid tumors, which unambiguously mapped to 3,107 unique Entrez Gene IDs, representative of all human chromosomes; however, by karyology, one of the hybrid tumors (GB-749) had a total of 15 human chromosomes in its cells. Among the genes mapped, 39 probe sets, representing 33 unique Entrez Gene IDs, complied with the detection criteria in all hybrid tumor samples. Five of these 33 genes encode transcription factors that are known to regulate cell growth and differentiation; five encode cell adhesion- and transmigration-associated proteins that participate in oncogenesis and/or metastasis and invasion; and additional genes encode proteins involved in signaling pathways, regulation of apoptosis, DNA repair, and multidrug resistance. These findings were corroborated by PCR and reverse transcription PCR, showing the presence of human alphoid (α)-satellite DNA and the F11R transcripts in additional tumor transplant generations. We posit that in-vivo fusion discloses genes implicated in tumor progression, and gene families coding for the organoid phenotype. Thus, cancer cells can transduce adjacent stromal cells, with the resulting progeny having permanently transcribed genes with malignant and other gene functions of the donor DNA. Using heterospecific in-vivo cell fusion, genes encoding oncogenic and organogenic traits may be identified.
Collapse
Affiliation(s)
- David M. Goldenberg
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, New Jersey, United States of America
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
- * E-mail:
| | - Robert J. Rooney
- Genome Explorations, Inc., Memphis, Tennessee, United States of America
| | - Meiyu Loo
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| | - Donglin Liu
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| | - Chien-Hsing Chang
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| |
Collapse
|
466
|
Leong TL, Marini KD, Rossello FJ, Jayasekara SN, Russell PA, Prodanovic Z, Kumar B, Ganju V, Alamgeer M, Irving LB, Steinfort DP, Peacock CD, Cain JE, Szczepny A, Watkins DN. Genomic characterisation of small cell lung cancer patient-derived xenografts generated from endobronchial ultrasound-guided transbronchial needle aspiration specimens. PLoS One 2014; 9:e106862. [PMID: 25191746 PMCID: PMC4156408 DOI: 10.1371/journal.pone.0106862] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/02/2014] [Indexed: 12/25/2022] Open
Abstract
Patient-derived xenograft (PDX) models generated from surgical specimens are gaining popularity as preclinical models of cancer. However, establishment of PDX lines from small cell lung cancer (SCLC) patients is difficult due to very limited amount of available biopsy material. We asked whether SCLC cells obtained from endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) could generate PDX lines that maintained the phenotypic and genetic characteristics of the primary tumor. Following successful EBUS-TBNA sampling for diagnostic purposes, we obtained an extra sample for cytologic analysis and implantation into the flanks of immunodeficient mice. Animals were monitored for engraftment for up to 6 months. Histopathologic and immunohistochemical analysis, and targeted next-generation re-sequencing, were then performed in both the primary sample and the derivative PDX line. A total of 12 patients were enrolled in the study. EBUS-TBNA aspirates yielded large numbers of viable tumor cells sufficient to inject between 18,750 and 1,487,000 cells per flank, and to yield microgram quantities of high-quality DNA. Of these, samples from 10 patients generated xenografts (engraftment rate 83%) with a mean latency of 104 days (range 63–188). All but one maintained a typical SCLC phenotype that closely matched the original sample. Identical mutations that are characteristic of SCLC were identified in both the primary sample and xenograft line. EBUS-TBNA has the potential to be a powerful tool in the development of new targeting strategies for SCLC patients by providing large numbers of viable tumor cells suitable for both xenografting and complex genomic analysis.
Collapse
Affiliation(s)
- Tracy L. Leong
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
| | - Kieren D. Marini
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
| | - Fernando J. Rossello
- Monash University, Clayton, Victoria, Australia
- Life Sciences Computation Centre, Victorian Life Sciences Computation Initiative, Carlton, Victoria, Australia
| | | | - Prudence A. Russell
- Department of Anatomical Pathology, St Vincent's Hospital, Fitzroy, Melbourne, Victoria, Australia
| | - Zdenka Prodanovic
- Department of Pathology, Monash Health, Clayton, Victoria, Australia
| | - Beena Kumar
- Department of Pathology, Monash Health, Clayton, Victoria, Australia
| | - Vinod Ganju
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
- Department of Medical Oncology, Monash Health, East Bentleigh, Victoria, Australia
| | - Muhammad Alamgeer
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
- Department of Medical Oncology, Monash Health, East Bentleigh, Victoria, Australia
| | - Louis B. Irving
- Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Daniel P. Steinfort
- Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Craig D. Peacock
- Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jason E. Cain
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
| | - Anette Szczepny
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
- * E-mail: (DNW); (AS)
| | - D. Neil Watkins
- MIMR-PHI Institute, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- * E-mail: (DNW); (AS)
| |
Collapse
|
467
|
Tignanelli CJ, Loeza SGH, Yeh JJ. KRAS and PIK3CA Mutation Frequencies in Patient-derived Xenograft Models of Pancreatic and Colorectal Cancer Are Reflective of Patient Tumors and Stable Across Passages. Am Surg 2014. [DOI: 10.1177/000313481408000920] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
One obstacle in the translation of advances in cancer research into the clinic is a deficiency of adequate preclinical models that recapitulate human disease. Patient-derived xenograft (PDX) models are established by engrafting patient tumor tissue into mice and are advantageous because they capture tumor heterogeneity. One concern with these models is that selective pressure could lead to mutational drift and thus be an inaccurate reflection of patient tumors. Therefore, we evaluated if mutational frequency in PDX models is reflective of patient populations and if crucial mutations are stable across passages. We examined KRAS and PIK3CA gene mutations from pancreatic ductal adenocarcinoma (PDAC) (n = 30) and colorectal cancer (CRC) (n = 37) PDXs for as many as eight passages. DNA was isolated from tumors and target sequences were amplified by polymerase chain reaction. KRAS codons 12/13 and PIK3CA codons 542/545/1047 were examined using pyrosequencing. Twenty-three of 30 (77%) PDAC PDXs had KRAS mutations and one of 30 (3%) had PIK3CA mutations. Fifteen of 37 (41%) CRC PDXs had KRAS mutations and three of 37 (8%) had PIK3CA mutations. Mutations were 100 per cent preserved across passages. We found that the frequency of KRAS (77%) and PIK3CA (3%) mutations in PDAC PDX was similar to frequencies in patient tumors (71 to 100% KRAS, 0 to 11% PIK3CA). Similarly, KRAS (41%) and PIK3CA (8%) mutations in CRC PDX closely paralleled patient tumors (35 to 51% KRAS, 12 to 21% PIK3CA). The accurate mirroring and stability of genetic changes in PDX models compared with patient tumors suggest that these models are good preclinical surrogates for patient tumors.
Collapse
Affiliation(s)
| | - Silvia G. Herrera Loeza
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Jen Jen Yeh
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
468
|
Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, Clarke RB, de Jong S, Jonkers J, Mælandsmo GM, Roman-Roman S, Seoane J, Trusolino L, Villanueva A. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov 2014; 4:998-1013. [PMID: 25185190 PMCID: PMC4167608 DOI: 10.1158/2159-8290.cd-14-0001] [Citation(s) in RCA: 1222] [Impact Index Per Article: 111.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED Recently, there has been an increasing interest in the development and characterization of patient-derived tumor xenograft (PDX) models for cancer research. PDX models mostly retain the principal histologic and genetic characteristics of their donor tumor and remain stable across passages. These models have been shown to be predictive of clinical outcomes and are being used for preclinical drug evaluation, biomarker identification, biologic studies, and personalized medicine strategies. This article summarizes the current state of the art in this field, including methodologic issues, available collections, practical applications, challenges and shortcomings, and future directions, and introduces a European consortium of PDX models. SIGNIFICANCE PDX models are increasingly used in translational cancer research. These models are useful for drug screening, biomarker development, and the preclinical evaluation of personalized medicine strategies. This review provides a timely overview of the key characteristics of PDX models and a detailed discussion of future directions in the field.
Collapse
Affiliation(s)
| | | | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow
| | | | | | | | - Robert B Clarke
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Jos Jonkers
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | | | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS; and Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Alberto Villanueva
- Catalan Institute of Oncology-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
469
|
Monsma DJ, Cherba DM, Richardson PJ, Vance S, Rangarajan S, Dylewski D, Eugster E, Scott SB, Beuschel NL, Davidson PJ, Axtell R, Mitchell D, Lester EP, Junewick JJ, Webb CP, Monks NR. Using a rhabdomyosarcoma patient-derived xenograft to examine precision medicine approaches and model acquired resistance. Pediatr Blood Cancer 2014; 61:1570-7. [PMID: 24687871 DOI: 10.1002/pbc.25039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/05/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND Precision (Personalized) medicine has the potential to revolutionize patient health care especially for many cancers where the fundamental disease etiology remains either elusive or has no available therapy. Here we outline a study in alveolar rhabdomyosarcoma, in which we use gene expression profiling and a series of drug prediction algorithms combined with a matched patient-derived xenograft (PDX) model to test bioinformatically predicted therapies. PROCEDURE A PDX model was developed from a patient biopsy and a number of drugs identified using gene expression analysis in combination with drug prediction algorithms. Drugs chosen from each of the predictive methodologies, along with the patient's standard-of-care therapy (ICE-T), were tested in vivo in the PDX tumor. A second study was initiated using the tumors that re-grew following the ICE-T treatment. Further expression analysis identified additional therapies with potential anti-tumor efficacy. RESULTS A number of the predicted therapies were found to be active against the tumors in particular BGJ398 (FGFR2) and ICE-T. Re-transplanted ICE-T treated tumorgrafts demonstrated a decreased response to ICE-T recapitulating the patient's refractory disease. Gene expression profiling of the ICE-T treated tumorgrafts identified cytarabine (SLC29A1) as a potential therapy, which was shown, along with BGJ398, to be highly active in vivo. CONCLUSIONS This study illustrates that PDX models are suitable surrogates for testing potential therapeutic strategies based on gene expression analysis, modeling clinical drug resistance and hold the potential to assist in guiding prospective patient care.
Collapse
Affiliation(s)
- David J Monsma
- Van Andel Research Institute, Center for Translational Medicine, Grand Rapids, Michigan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
470
|
Tignanelli CJ, Loeza SGH, Yeh JJ. KRAS and PIK3CA mutation frequencies in patient-derived xenograft models of pancreatic and colorectal cancer are reflective of patient tumors and stable across passages. Am Surg 2014; 80:873-877. [PMID: 25197873 PMCID: PMC4425299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
One obstacle in the translation of advances in cancer research into the clinic is a deficiency of adequate preclinical models that recapitulate human disease. Patient-derived xenograft (PDX) models are established by engrafting patient tumor tissue into mice and are advantageous because they capture tumor heterogeneity. One concern with these models is that selective pressure could lead to mutational drift and thus be an inaccurate reflection of patient tumors. Therefore, we evaluated if mutational frequency in PDX models is reflective of patient populations and if crucial mutations are stable across passages. We examined KRAS and PIK3CA gene mutations from pancreatic ductal adenocarcinoma (PDAC) (n = 30) and colorectal cancer (CRC) (n = 37) PDXs for as many as eight passages. DNA was isolated from tumors and target sequences were amplified by polymerase chain reaction. KRAS codons 12/13 and PIK3CA codons 542/545/1047 were examined using pyrosequencing. Twenty-three of 30 (77%) PDAC PDXs had KRAS mutations and one of 30 (3%) had PIK3CA mutations. Fifteen of 37 (41%) CRC PDXs had KRAS mutations and three of 37 (8%) had PIK3CA mutations. Mutations were 100 per cent preserved across passages. We found that the frequency of KRAS (77%) and PIK3CA (3%) mutations in PDAC PDX was similar to frequencies in patient tumors (71 to 100% KRAS, 0 to 11% PIK3CA). Similarly, KRAS (41%) and PIK3CA (8%) mutations in CRC PDX closely paralleled patient tumors (35 to 51% KRAS, 12 to 21% PIK3CA). The accurate mirroring and stability of genetic changes in PDX models compared with patient tumors suggest that these models are good preclinical surrogates for patient tumors.
Collapse
Affiliation(s)
| | | | - Jen Jen Yeh
- Department of Surgery, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
471
|
Denayer T, Stöhr T, Roy MV. Animal models in translational medicine: Validation and prediction. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.nhtm.2014.08.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
472
|
Brand TM, Iida M, Stein AP, Corrigan KL, Braverman CM, Luthar N, Toulany M, Gill PS, Salgia R, Kimple RJ, Wheeler DL. AXL mediates resistance to cetuximab therapy. Cancer Res 2014; 74:5152-64. [PMID: 25136066 DOI: 10.1158/0008-5472.can-14-0294] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The EGFR antibody cetuximab is used to treat numerous cancers, but intrinsic and acquired resistance to this agent is a common clinical outcome. In this study, we show that overexpression of the oncogenic receptor tyrosine kinase AXL is sufficient to mediate acquired resistance to cetuximab in models of non-small cell lung cancer (NSCLC) and head and neck squamous cell carcinoma (HNSCC), where AXL was overexpressed, activated, and tightly associated with EGFR expression in cells resistant to cetuximab (Ctx(R) cells). Using RNAi methods and novel AXL-targeting agents, we found that AXL activation stimulated cell proliferation, EGFR activation, and MAPK signaling in Ctx(R) cells. Notably, EGFR directly regulated the expression of AXL mRNA through MAPK signaling and the transcription factor c-Jun in Ctx(R) cells, creating a positive feedback loop that maintained EGFR activation by AXL. Cetuximab-sensitive parental cells were rendered resistant to cetuximab by stable overexpression of AXL or stimulation with EGFR ligands, the latter of which increased AXL activity and association with the EGFR. In tumor xenograft models, the development of resistance following prolonged treatment with cetuximab was associated with AXL hyperactivation and EGFR association. Furthermore, in an examination of patient-derived xenografts established from surgically resected HNSCCs, AXL was overexpressed and activated in tumors that displayed intrinsic resistance to cetuximab. Collectively, our results identify AXL as a key mediator of cetuximab resistance, providing a rationale for clinical evaluation of AXL-targeting drugs to treat cetuximab-resistant cancers. Cancer Res; 74(18); 5152-64. ©2014 AACR.
Collapse
Affiliation(s)
- Toni M Brand
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Andrew P Stein
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kelsey L Corrigan
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Cara M Braverman
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Neha Luthar
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Parkash S Gill
- Departments of Medicine and Pathology, University of Southern California, Los Angeles, California
| | - Ravi Salgia
- Department of Medicine, Division of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
473
|
McNeill RS, Schmid RS, Bash RE, Vitucci M, White KK, Werneke AM, Constance BH, Huff B, Miller CR. Modeling astrocytoma pathogenesis in vitro and in vivo using cortical astrocytes or neural stem cells from conditional, genetically engineered mice. J Vis Exp 2014:e51763. [PMID: 25146643 DOI: 10.3791/51763] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Current astrocytoma models are limited in their ability to define the roles of oncogenic mutations in specific brain cell types during disease pathogenesis and their utility for preclinical drug development. In order to design a better model system for these applications, phenotypically wild-type cortical astrocytes and neural stem cells (NSC) from conditional, genetically engineered mice (GEM) that harbor various combinations of floxed oncogenic alleles were harvested and grown in culture. Genetic recombination was induced in vitro using adenoviral Cre-mediated recombination, resulting in expression of mutated oncogenes and deletion of tumor suppressor genes. The phenotypic consequences of these mutations were defined by measuring proliferation, transformation, and drug response in vitro. Orthotopic allograft models, whereby transformed cells are stereotactically injected into the brains of immune-competent, syngeneic littermates, were developed to define the role of oncogenic mutations and cell type on tumorigenesis in vivo. Unlike most established human glioblastoma cell line xenografts, injection of transformed GEM-derived cortical astrocytes into the brains of immune-competent littermates produced astrocytomas, including the most aggressive subtype, glioblastoma, that recapitulated the histopathological hallmarks of human astrocytomas, including diffuse invasion of normal brain parenchyma. Bioluminescence imaging of orthotopic allografts from transformed astrocytes engineered to express luciferase was utilized to monitor in vivo tumor growth over time. Thus, astrocytoma models using astrocytes and NSC harvested from GEM with conditional oncogenic alleles provide an integrated system to study the genetics and cell biology of astrocytoma pathogenesis in vitro and in vivo and may be useful in preclinical drug development for these devastating diseases.
Collapse
Affiliation(s)
- Robert S McNeill
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine
| | - Ralf S Schmid
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine
| | - Ryan E Bash
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine
| | - Mark Vitucci
- Curriculum in Genetics and Molecular Biology, University of North Carolina School of Medicine
| | - Kristen K White
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine
| | - Andrea M Werneke
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine
| | - Brian H Constance
- Biological and Biomedical Sciences Program, University of North Carolina School of Medicine
| | - Byron Huff
- Department of Radiation Oncology, Emory University School of Medicine
| | - C Ryan Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine; Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine; Department of Neurology, Neurosciences Center, University of North Carolina School of Medicine;
| |
Collapse
|
474
|
Fleet JC. Animal models of gastrointestinal and liver diseases. New mouse models for studying dietary prevention of colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2014; 307:G249-59. [PMID: 24875098 PMCID: PMC4121636 DOI: 10.1152/ajpgi.00019.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is a heterogeneous disease that is one of the major causes of cancer death in the U.S. There is evidence that lifestyle factors like diet can modulate the course of this disease. Demonstrating the benefit and mechanism of action of dietary interventions against colon cancer will require studies in preclinical models. Many mouse models have been developed to study colon cancer but no single model can reflect all types of colon cancer in terms of molecular etiology. In addition, many models develop only low-grade cancers and are confounded by development of the disease outside of the colon. This review will discuss how mice can be used to model human colon cancer and it will describe a variety of new mouse models that develop colon-restricted cancer as well as more advanced phenotypes for studies of late-state disease.
Collapse
Affiliation(s)
- James C. Fleet
- 1Department of Nutrition Science, Purdue University, West Lafayette, Indiana; and ,2Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
475
|
Chonghaile TN, Roderick JE, Glenfield C, Ryan J, Sallan SE, Silverman LB, Loh ML, Hunger SP, Wood B, DeAngelo DJ, Stone R, Harris M, Gutierrez A, Kelliher MA, Letai A. Maturation stage of T-cell acute lymphoblastic leukemia determines BCL-2 versus BCL-XL dependence and sensitivity to ABT-199. Cancer Discov 2014; 4:1074-87. [PMID: 24994123 DOI: 10.1158/2159-8290.cd-14-0353] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Acute lymphoblastic leukemia (ALL) is a hematopoietic malignancy derived from immature B-lymphoid and T-lymphoid cells (T-ALL). In T-ALL, there is an early T-cell progenitor (ETP) subgroup that has a very high risk for relapse. In this study, we used mitochondrial BH3 profiling to determine antiapoptotic protein dependencies in T-ALL. We found that T-ALL cell lines and primary patient samples are dependent upon BCL-XL, except when the cancer bears an ETP phenotype, in which case it is BCL-2 dependent. These distinctions directly relate to differential sensitivity to the BH3 mimetics ABT-263 and ABT-199, both in vitro and in vivo. We thus describe for the first time a change of antiapoptotic protein dependence that is related to the differentiation stage of the leukemic clone. Our findings demonstrate that BCL-2 is a clinically relevant target for therapeutic intervention with ABT-199 in ETP-ALL. SIGNIFICANCE ETP T-ALL is a treatment-resistant subtype of T-ALL for which novel targeted therapies are urgently needed. We have discovered, through BH3 profiling, that ETP-ALL is BCL-2 dependent and is very sensitive to in vitro and in vivo treatment with ABT-199, a drug well tolerated in clinical trials.
Collapse
Affiliation(s)
- Triona Ni Chonghaile
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Justine E Roderick
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Cian Glenfield
- Department of Genetics, The Smurfit Institute, Trinity College, Dublin, Ireland
| | - Jeremy Ryan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Stephen E Sallan
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Lewis B Silverman
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, California
| | - Stephen P Hunger
- Pediatric Hematology/Oncology/BMT, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado
| | - Brent Wood
- Department of Medicine, University of Washington, Seattle, Washington
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Richard Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marian Harris
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Alejandro Gutierrez
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michelle A Kelliher
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
476
|
Obesity and resistance to cancer chemotherapy: interacting roles of inflammation and metabolic dysregulation. Clin Pharmacol Ther 2014; 96:458-63. [PMID: 24960521 DOI: 10.1038/clpt.2014.136] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
Abstract
The prevalence of obesity, an established risk factor for many chronic diseases, including several types of cancer, has risen steadily over the past four decades in the United States and worldwide. To date, research in this area has focused on the epidemiologic associations between obesity and cancer risk, as well as on the mechanisms underlying those associations. However, an emerging but understudied issue of clinical importance is the diminution of chemotherapeutic efficacy in obese cancer patients. The mechanisms underlying the negative impact of obesity on therapeutic responses are likely multifactorial. The effects of obesity on chemotherapy drug pharmacokinetics and dosage have been extensively reviewed elsewhere, so this review will focus on the interplay among obesity, increased inflammation, metabolic perturbations, and chemoresistance. The ultimate goal of this review is to delineate areas for future research that could lead to the identification of new targets and strategies for improved cancer outcomes in obese patients.
Collapse
|
477
|
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: 124] [Impact Index Per Article: 11.3] [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
|
478
|
Kung PP, Martinez R, Zhu Z, Zager M, Blasina A, Rymer I, Hallin J, Xu M, Carroll C, Chionis J, Wells P, Kozminski K, Fan J, Guicherit O, Huang B, Cui M, Liu C, Huang Z, Sistla A, Yang J, Murray BW. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol Cancer Ther 2014; 13:2104-15. [PMID: 24928852 DOI: 10.1158/1535-7163.mct-14-0083-t] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Breast cancer patients with tumors lacking the three diagnostic markers (ER, PR, and HER2) are classified as triple-negative (primarily basal-like) and have poor prognosis because there is no disease-specific therapy available. To address this unmet medical need, gene expression analyses using more than a thousand breast cancer samples were conducted, which identified elevated centromere protein E (CENP-E) expression in the basal-a molecular subtype relative to other subtypes. CENP-E, a mitotic kinesin component of the spindle assembly checkpoint, is shown to be induced in basal-a tumor cell lines by the mitotic spindle inhibitor drug docetaxel. CENP-E knockdown by inducible shRNA reduces basal-a breast cancer cell viability. A potent, selective CENP-E inhibitor (PF-2771) was used to define the contribution of CENP-E motor function to basal-like breast cancer. Mechanistic evaluation of PF-2771 in basal-a tumor cells links CENP-E-dependent molecular events (e.g., phosphorylation of histone H3 Ser-10; phospho-HH3-Ser10) to functional outcomes (e.g., chromosomal congression defects). Across a diverse panel of breast cell lines, CENP-E inhibition by PF-2771 selectively inhibits proliferation of basal breast cancer cell lines relative to premalignant ones and its response correlates with the degree of chromosomal instability. Pharmacokinetic-pharmacodynamic efficacy analysis in a basal-a xenograft tumor model shows that PF-2771 exposure is well correlated with increased phospho-HH3-Ser10 levels and tumor growth regression. Complete tumor regression is observed in a patient-derived, basal-a breast cancer xenograft tumor model treated with PF-2771. Tumor regression is also observed with PF-2771 in a taxane-resistant basal-a model. Taken together, CENP-E may be an effective therapeutic target for patients with triple-negative/basal-a breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Anand Sistla
- Pharmaceuticals Science, Pfizer Worldwide Research and Development, La Jolla Laboratories, San Diego, California
| | | | | |
Collapse
|
479
|
Chicaybam L, Bonamino MH. Moving Receptor Redirected Adoptive Cell Therapy Toward Fine Tuning of Antitumor Responses. Int Rev Immunol 2014; 33:402-16. [DOI: 10.3109/08830185.2014.917412] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
480
|
Herrmann D, Conway JRW, Vennin C, Magenau A, Hughes WE, Morton JP, Timpson P. Three-dimensional cancer models mimic cell-matrix interactions in the tumour microenvironment. Carcinogenesis 2014; 35:1671-9. [PMID: 24903340 DOI: 10.1093/carcin/bgu108] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Basic in vitro systems can be used to model and assess complex diseases, such as cancer. Recent advances in this field include the incorporation of multiple cell types and extracellular matrix proteins into three-dimensional (3D) models to recapitulate the structure, organization and functionality of live tissue in situ. Cells within such a 3D environment behave very differently from cells on two-dimensional (2D) substrates, as cell-matrix interactions trigger signalling pathways and cellular responses in 3D, which may not be observed in 2D. Thus, the use of 3D systems can be advantageous for the assessment of disease progression over 2D set-ups alone. Here, we highlight the current advantages and challenges of employing 3D systems in the study of cancer and provide an overview to guide the appropriate use of distinct models in cancer research.
Collapse
Affiliation(s)
- David Herrmann
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - James R W Conway
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Astrid Magenau
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - William E Hughes
- Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and
| | - Jennifer P Morton
- The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| |
Collapse
|
481
|
Clark AM, Wheeler SE, Taylor DP, Pillai VC, Young CL, Prantil-Baun R, Nguyen T, Stolz DB, Borenstein JT, Lauffenburger DA, Venkataramanan R, Griffith LG, Wells A. A microphysiological system model of therapy for liver micrometastases. Exp Biol Med (Maywood) 2014; 239:1170-9. [PMID: 24821820 DOI: 10.1177/1535370214532596] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Metastasis accounts for almost 90% of cancer-associated mortality. The effectiveness of cancer therapeutics is limited by the protective microenvironment of the metastatic niche and consequently these disseminated tumors remain incurable. Metastatic disease progression continues to be poorly understood due to the lack of appropriate model systems. To address this gap in understanding, we propose an all-human microphysiological system that facilitates the investigation of cancer behavior in the liver metastatic niche. This existing LiverChip is a 3D-system modeling the hepatic niche; it incorporates a full complement of human parenchymal and non-parenchymal cells and effectively recapitulates micrometastases. Moreover, this system allows real-time monitoring of micrometastasis and assessment of human-specific signaling. It is being utilized to further our understanding of the efficacy of chemotherapeutics by examining the activity of established and novel agents on micrometastases under conditions replicating diurnal variations in hormones, nutrients and mild inflammatory states using programmable microdispensers. These inputs affect the cues that govern tumor cell responses. Three critical signaling groups are targeted: the glucose/insulin responses, the stress hormone cortisol and the gut microbiome in relation to inflammatory cues. Currently, the system sustains functioning hepatocytes for a minimum of 15 days; confirmed by monitoring hepatic function (urea, α-1-antitrypsin, fibrinogen, and cytochrome P450) and injury (AST and ALT). Breast cancer cell lines effectively integrate into the hepatic niche without detectable disruption to tissue, and preliminary evidence suggests growth attenuation amongst a subpopulation of breast cancer cells. xMAP technology combined with systems biology modeling are also employed to evaluate cellular crosstalk and illustrate communication networks in the early microenvironment of micrometastases. This model is anticipated to identify new therapeutic strategies for metastasis by elucidating the paracrine effects between the hepatic and metastatic cells, while concurrently evaluating agent efficacy for metastasis, metabolism and tolerability.
Collapse
Affiliation(s)
- Amanda M Clark
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Sarah E Wheeler
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Donald P Taylor
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Venkateswaran C Pillai
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Carissa L Young
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | | | - Transon Nguyen
- Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| | - Donna B Stolz
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | | | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | - Raman Venkataramanan
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02319, USA
| | - Alan Wells
- Departments of Pathology, Cell Biology, Pharmaceutical Sciences, and Bioengineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh and Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
| |
Collapse
|
482
|
Guerrero-Preston R. Avatars of personalized cancer therapy can lead to a reduction in head and neck cancer survival disparities. Per Med 2014; 11:243-246. [DOI: 10.2217/pme.14.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
483
|
Fong ELS, Martinez M, Yang J, Mikos AG, Navone NM, Harrington DA, Farach-Carson MC. Hydrogel-based 3D model of patient-derived prostate xenograft tumors suitable for drug screening. Mol Pharm 2014; 11:2040-50. [PMID: 24779589 PMCID: PMC4096229 DOI: 10.1021/mp500085p] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The lack of effective
therapies for bone metastatic prostate cancer
(PCa) underscores the need for accurate models of the disease to enable
the discovery of new therapeutic targets and to test drug sensitivities
of individual tumors. To this end, the patient-derived xenograft (PDX)
PCa model using immunocompromised mice was established to model the
disease with greater fidelity than is possible with currently employed
cell lines grown on tissue culture plastic. However, poorly adherent
PDX tumor cells exhibit low viability in standard culture, making
it difficult to manipulate these cells for subsequent controlled mechanistic
studies. To overcome this challenge, we encapsulated PDX tumor cells
within a three-dimensional hyaluronan-based hydrogel and demonstrated
that the hydrogel maintains PDX cell viability with continued native
androgen receptor expression. Furthermore, a differential sensitivity
to docetaxel, a chemotherapeutic drug, was observed as compared to
a traditional PCa cell line. These findings underscore the potential
impact of this novel 3D PDX PCa model as a diagnostic platform for
rapid drug evaluation and ultimately push personalized medicine toward
clinical reality.
Collapse
Affiliation(s)
- Eliza L S Fong
- Departments of Biochemistry and Cell Biology and ‡Bioengineering, Rice University , Houston, Texas 77005, United States
| | | | | | | | | | | | | |
Collapse
|
484
|
Abstract
Despite the millions of dollars spent on target validation and drug optimization in preclinical models, most therapies still fail in phase III clinical trials. Our current model systems, or the way we interpret data from them, clearly do not have sufficient clinical predictive power. Current opinion suggests that this is because the cell lines and xenografts that are commonly used are inadequate models that do not effectively mimic and predict human responses. This has become such a widespread belief that it approaches dogma in the field of drug discovery and optimization and has spurred a surge in studies devoted to the development of more sophisticated animal models such as orthotopic patient-derived xenografts in an attempt to obtain more accurate estimates of whether particular cancers will respond to given treatments. Here, we explore the evidence that has led to the move away from the use of in vitro cell lines and toward various forms of xenograft models for drug screening and development. We review some of the pros and cons of each model and give an overview of ways in which the use of cell lines could be modified to improve the predictive capacity of this well-defined model.
Collapse
Affiliation(s)
- Jennifer L Wilding
- Authors' Affiliation: Department of Oncology, Cancer and Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | | |
Collapse
|
485
|
Khaled WT, Liu P. Cancer mouse models: past, present and future. Semin Cell Dev Biol 2014; 27:54-60. [PMID: 24718321 DOI: 10.1016/j.semcdb.2014.04.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/29/2014] [Accepted: 04/01/2014] [Indexed: 12/26/2022]
Abstract
The development and advances in gene targeting technology over the past three decades has facilitated the generation of cancer mouse models that recapitulate features of human malignancies. These models have been and still remain instrumental in revealing the complexities of human cancer biology. However, they will need to evolve in the post-genomic era of cancer research. In this review we will highlight some of the key developments over the past decades and will discuss the new possibilities of cancer mouse models in the light of emerging powerful gene manipulating tools.
Collapse
Affiliation(s)
- Walid T Khaled
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK.
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK.
| |
Collapse
|
486
|
Romano RA, Sinha S. Family matters: sibling rivalry and bonding between p53 and p63 in cancer. Exp Dermatol 2014; 23:238-9. [DOI: 10.1111/exd.12356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Rose-Anne Romano
- Department of Biochemistry; State University of New York at Buffalo; Buffalo NY USA
| | - Satrajit Sinha
- Department of Biochemistry; State University of New York at Buffalo; Buffalo NY USA
| |
Collapse
|
487
|
Das Thakur M, Pryer NK, Singh M. Mouse tumour models to guide drug development and identify resistance mechanisms. J Pathol 2014; 232:103-11. [PMID: 24122209 DOI: 10.1002/path.4285] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 12/11/2022]
Abstract
We need improved, translatable and predictive tumour models for the evaluation of response and the evolution of resistance to targeted therapeutics. We provide a review of the use of different types of preclinical tumour models to evaluate novel anticancer agents, and model the rapidly evolving landscape of resistance to targeted therapy. We focus on describing the various preclinical models available for candidate drug development and design considerations for preclinical experiments, depending on the aspect of drug action being interrogated. We discuss selected examples of how experimental findings have translated into clinical outcomes for targeted agents, predicted mechanisms that drive resistance and strategies to overcome the evolution thereof. We discuss challenges in preclinical experimental design and interpretation and possible improvements in animal models of therapeutic response and resistance, with an emphasis on improved translation of experimental research into clinical practice.
Collapse
Affiliation(s)
- Meghna Das Thakur
- Oncology Pharmacology, Novartis Institutes for Biomedical Research, Emeryville, CA, USA
| | | | | |
Collapse
|
488
|
Xin H, Wang K, Hu G, Xie F, Ouyang K, Tang X, Wang M, Wen D, Zhu Y, Qin X. Establishment and characterization of 7 novel hepatocellular carcinoma cell lines from patient-derived tumor xenografts. PLoS One 2014; 9:e85308. [PMID: 24416385 PMCID: PMC3887059 DOI: 10.1371/journal.pone.0085308] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/25/2013] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common cancer with poor prognosis worldwide and the molecular mechanism is not well understood. This study aimed to establish a collection of human HCC cell lines from patient-derived xenograft (PDX) models. From the 20 surgical HCC sample collections, 7 tumors were successfully developed in immunodeficient mice and further established 7 novel HCC cell lines (LIXC002, LIXC003, LIXC004, LIXC006, LIXC011, LIXC012 and CPL0903) by primary culture. The characterization of cell lines was defined by morphology, growth kinetics, cell cycle, chromosome analysis, short tandem repeat (STR) analysis, molecular profile, and tumorigenicity. Additionally, response to clinical chemotherapeutics was validated both in vitro and in vivo. STR analysis indicated that all cell lines were unique cells different from known cell lines and free of contamination by bacteria or mycoplasma. The other findings were quite heterogeneous between individual lines. Chromosome aberration could be found in all cell lines. Alpha-fetoprotein was overexpressed only in 3 out of 7 cell lines. 4 cell lines expressed high level of vimentin. Ki67 was strongly stained in all cell lines. mRNA level of retinoic acid induced protein 3 (RAI3) was decreased in all cell lines. The 7 novel cell lines showed variable sensitivity to 8 tested compounds. LIXC011 and CPL0903 possessed multiple drug resistance property. Sorafenib inhibited xenograft tumor growth of LIXC006, but not of LIXC012. Our results indicated that the 7 novel cell lines with low passage maintaining their clinical and pathological characters could be good tools for further exploring the molecular mechanism of HCC and anti-cancer drug screening.
Collapse
Affiliation(s)
- Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Ke Wang
- Shanghai ChemPartner Co., LTD, Shanghai, China
| | - Gang Hu
- Shanghai ChemPartner Co., LTD, Shanghai, China
| | - Fubo Xie
- Shanghai ChemPartner Co., LTD, Shanghai, China
| | | | - Xuzhen Tang
- Shanghai ChemPartner Co., LTD, Shanghai, China
| | - Minjun Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Danyi Wen
- Shanghai ChemPartner Co., LTD, Shanghai, China
| | - Yizhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoran Qin
- Shanghai ChemPartner Co., LTD, Shanghai, China
| |
Collapse
|
489
|
Wartha K, Herting F, Hasmann M. Fit-for purpose use of mouse models to improve predictivity of cancer therapeutics evaluation. Pharmacol Ther 2014; 142:351-61. [PMID: 24412280 DOI: 10.1016/j.pharmthera.2014.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
UNLABELLED Preclinical animal models are useful tools to better understand tumor initiation and progression and to predict the activity of an anticancer agent in the clinic. Ideally, these models should recapitulate the biological characteristics of the tumor and of the related tumor microenvironment (e.g. vasculature, immune cells) in patients. Even if several examples of translational success have been reported it is a matter of fact that clinical trials in oncology often fail to meet their primary endpoints despite encouraging preclinical data. For this reason, there is an increasing need of improved and more predictive models. This review aims to give an overview on existing mouse models for preclinical evaluation of cancer therapeutics and their applicability. Different types of mouse models commonly used for the evaluation of cancer therapeutics are described and considerations for a "fit-for purpose" application of these models for the evaluation of different cancer therapeutics dependent on their mode of action are outlined. Furthermore, considerations for study design and data interpretation to translatability of findings into the clinics are given. CONCLUSION Detailed knowledge of the molecular/biological properties of the respective model, diligent experimental setup, and awareness of its limitations are indispensable prerequisites for the successful translational use of animal models.
Collapse
Affiliation(s)
- K Wartha
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany.
| | - F Herting
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - M Hasmann
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
490
|
Even-Desrumeaux K, Nevoltris D, Lavaut MN, Alim K, Borg JP, Audebert S, Kerfelec B, Baty D, Chames P. Masked selection: a straightforward and flexible approach for the selection of binders against specific epitopes and differentially expressed proteins by phage display. Mol Cell Proteomics 2013; 13:653-65. [PMID: 24361863 DOI: 10.1074/mcp.o112.025486] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Phage display is a well-established procedure to isolate binders against a wide variety of antigens that can be performed on purified antigens, but also on intact cells. As selection steps are performed in vitro, it is possible to focus the outcome of the selection on relevant epitopes by performing some additional steps, such as depletion or competitive elutions. However in practice, the efficiency of these steps is often limited and can lead to inconsistent results. We have designed a new selection method named masked selection, based on the blockade of unwanted epitopes to favor the targeting of relevant ones. We demonstrate the efficiency and flexibility of this method by selecting single-domain antibodies against a specific portion of a fusion protein, by selecting binders against several members of the seven transmembrane receptor family using transfected HEK cells, or by selecting binders against unknown breast cancer markers not expressed on normal samples. The relevance of this approach for antibody-based therapies was further validated by the identification of four of these markers, Epithelial cell adhesion molecule, Transferrin receptor 1, Metastasis cell adhesion molecule, and Sushi containing domain 2, using immunoprecipitation and mass spectrometry. This new phage display strategy can be applied to any type of antibody fragments or alternative scaffolds, and is especially suited for the rapid discovery and identification of cell surface markers.
Collapse
|
491
|
Seol HS, Kang HJ, Kang H, Lee SI, Kim NE, Kim TI, Chun SM, Kim TW, Yu CS, Suh YA, Singh SR, Chang S, Jang SJ. Development and characterization of a colon PDX model that reproduces drug responsiveness and the mutation profiles of its original tumor. Cancer Lett 2013; 345:56-64. [PMID: 24333725 DOI: 10.1016/j.canlet.2013.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/17/2013] [Accepted: 11/21/2013] [Indexed: 12/16/2022]
Abstract
Cultures of primary tumors are very useful as a personalized screening system for effective therapeutic options. We here describe an effective method of reproducing human primary colon tumors through primary culture and a mouse xenograft model. A total of 199 primary colon tumor cultures were successfully established under optimized conditions to enrich for tumor cells and to expand it for long-term storage in liquid nitrogen. To examine whether these stored cultures retained original tumor properties, fifty primary cultures were xenografted into NOD-SCID mouse. Histological and tumor marker analysis of four representative tumor xenografts revealed that all of the xenograft retained its primary tumor characteristics. Oncomap analysis further showed no change in the major mutations in the xenografts, confirming that our method faithfully reproduced human colon tumors. A drug sensitivity assay revealed that two of the primary cultures were hypersensitive to oxaliplatin rather than 5-FU, which was used in the patients, suggesting it as an effective therapeutic option. We thus present an effective, reproducible preclinical model for testing various personalized therapeutic options in colon cancer patients.
Collapse
Affiliation(s)
- Hyang Sook Seol
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | | | - Hyojeong Kang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Seul-I Lee
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Na Eun Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Tae Im Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Sung Min Chun
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Tae Won Kim
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Chang Sik Yu
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Young-Ah Suh
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| | - Suhwan Chang
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.
| | - Se Jin Jang
- Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.
| |
Collapse
|
492
|
Charafe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski J, Finetti P, Josselin E, Adelaide J, Nguyen TT, Monville F, Jacquemier J, Thomassin-Piana J, Pinna G, Jalaguier A, Lambaudie E, Houvenaeghel G, Xerri L, Harel-Bellan A, Chaffanet M, Viens P, Birnbaum D. ALDH1-positive cancer stem cells predict engraftment of primary breast tumors and are governed by a common stem cell program. Cancer Res 2013; 73:7290-300. [PMID: 24142344 DOI: 10.1158/0008-5472.can-12-4704] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer stem-like cells (CSC) have been widely studied, but their clinical relevance has yet to be established in breast cancer. Here, we report the establishment of primary breast tumor-derived xenografts (PDX) that encompass the main diversity of human breast cancer and retain the major clinicopathologic features of primary tumors. Successful engraftment was correlated with the presence of ALDH1-positive CSCs, which predicted prognosis in patients. The xenografts we developed showed a hierarchical cell organization of breast cancer with the ALDH1-positive CSCs constituting the tumorigenic cell population. Analysis of gene expression from functionally validated CSCs yielded a breast CSC signature and identified a core transcriptional program of 19 genes shared with murine embryonic, hematopoietic, and neural stem cells. This generalized stem cell program allowed the identification of potential CSC regulators, which were related mainly to metabolic processes. Using an siRNA genetic screen designed to target the 19 genes, we validated the functional role of this stem cell program in the regulation of breast CSC biology. Our work offers a proof of the functional importance of CSCs in breast cancer, and it establishes the reliability of PDXs for use in developing personalized CSC therapies for patients with breast cancer.
Collapse
Affiliation(s)
- Emmanuelle Charafe-Jauffret
- Authors' Affiliations: INSERM, U1068, CRCM, Molecular Oncology, Institut Paoli-Calmettes, Biopathology, Aix-Marseille Univ, UM 105, F-13284, Départements d'Oncologie Médicale, Chirugie oncologique, and Radiologie, Institut Paoli-Calmettes, Marseille; and Plateforme ARN interference PArI, CEA SACLAY, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
493
|
Dey N, Sun Y, Leyland-Jones B, De P. Evolution of Tumor Model: From Animal Model of Tumor to Tumor Model in Animal. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.49168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|