Academic literature on the topic 'Genetically-engineered mouse (GEM) models'
Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles
Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Genetically-engineered mouse (GEM) models.'
Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.
You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.
Journal articles on the topic "Genetically-engineered mouse (GEM) models"
Cho, Kyungjoo, Simon Weonsang Ro, Sang Hyun Seo, Youjin Jeon, Hyuk Moon, Do Young Kim, and Seung Up Kim. "Genetically Engineered Mouse Models for Liver Cancer." Cancers 12, no. 1 (December 19, 2019): 14. http://dx.doi.org/10.3390/cancers12010014.
Full textVarticovski, L., M. G. Hollingshead, M. R. Anver, A. I. Robles, J. E. Green, K. W. Hunter, G. Merlino, et al. "Preclinical testing using tumors from genetically engineered mouse mammary models." Journal of Clinical Oncology 24, no. 18_suppl (June 20, 2006): 10067. http://dx.doi.org/10.1200/jco.2006.24.18_suppl.10067.
Full textShibata, Maho, and Michael M. Shen. "Stem cells in genetically-engineered mouse models of prostate cancer." Endocrine-Related Cancer 22, no. 6 (September 4, 2015): T199—T208. http://dx.doi.org/10.1530/erc-15-0367.
Full textSzabova, Ludmila, Baktiar Karim, Melanie Gordon, Lucy Lu, Nathan Pate, and Zoe Weaver Ohler. "A Transplantable Syngeneic Allograft Mouse Model for Nongestational Choriocarcinoma of the Ovary." Veterinary Pathology 56, no. 3 (January 13, 2019): 399–403. http://dx.doi.org/10.1177/0300985818823669.
Full textGarcia, Patrick L., Aubrey L. Miller, and Karina J. Yoon. "Patient-Derived Xenograft Models of Pancreatic Cancer: Overview and Comparison with Other Types of Models." Cancers 12, no. 5 (May 22, 2020): 1327. http://dx.doi.org/10.3390/cancers12051327.
Full textFerri-Borgogno, Sammy, Sugata Barui, Amberly M. McGee, Tamara Griffiths, Pankaj K. Singh, Cortt G. Piett, Bidyut Ghosh, et al. "Paradoxical Role of AT-rich Interactive Domain 1A in Restraining Pancreatic Carcinogenesis." Cancers 12, no. 9 (September 21, 2020): 2695. http://dx.doi.org/10.3390/cancers12092695.
Full textDonson, Andrew, Kent Riemondy, Sujatha Venkataraman, Ahmed Gilani, Bridget Sanford, Andrea Griesinger, Vladimir Amani, et al. "MBRS-46. CHARTING NEOPLASTIC AND IMMUNE CELL HETEROGENEITY IN HUMAN AND GEM MODELS OF MEDULLOBLASTOMA USING scRNAseq." Neuro-Oncology 22, Supplement_3 (December 1, 2020): iii406. http://dx.doi.org/10.1093/neuonc/noaa222.555.
Full textMohammed, Altaf, Naveena B. Janakiram, Venkateshwar Madka, Min Li, Adam S. Asch, and Chinthalapally V. Rao. "Current Challenges and Opportunities for Chemoprevention of Pancreatic Cancer." Current Medicinal Chemistry 25, no. 22 (July 4, 2018): 2535–44. http://dx.doi.org/10.2174/0929867324666170209104453.
Full textPark, Jun Won, Hyejin Um, Hanna Yang, Joo Young Cha, Kyoung-June Lee, and Hark K. Kim. "CWP232291, a Wnt/β-catenin inhibitor, to suppress the growth and development of gastrointestinal cancers." Journal of Clinical Oncology 35, no. 15_suppl (May 20, 2017): e15534-e15534. http://dx.doi.org/10.1200/jco.2017.35.15_suppl.e15534.
Full textDe Velasco, Marco A., and Hirotsugu Uemura. "Preclinical Remodeling of Human Prostate Cancer through the PTEN/AKT Pathway." Advances in Urology 2012 (2012): 1–12. http://dx.doi.org/10.1155/2012/419348.
Full textDissertations / Theses on the topic "Genetically-engineered mouse (GEM) models"
Creedon, Helen. "Use of genetically engineered mouse models in preclinical drug development." Thesis, University of Edinburgh, 2015. http://hdl.handle.net/1842/15911.
Full textVallerand, David. "Etude du stroma de tumeurs mammaires humaines xénogreffées et de modèles transgéniques murins." Thesis, Paris 11, 2014. http://www.theses.fr/2014PA11T001.
Full textTumor development is a multi-step process influencing by interactions between tumor cells and surrounding stroma. Breast cancer development involves a high level of communication between mammary epithelial cells, inflammatory cells, myofibroblasts and endothelial cells. So, the tumoral microenvironment appears as a prime target for anti-tumoral treatment. The use of preclinical models is a critical step in development and validation processes of new therapies. Nevertheless, the role of stroma in these models is poorly understood.In order to evaluate stromal cell populations in breast cancer preclinical models, we combined flow cytometry analysis and immunohistochemistry to identify, and then quantify, various stromal populations as hematopoietic cells (lymphocytes, monocytes/macrophages, polymorphonuclear leukocytes) and non-hematopoietic cells (myofibroblasts, endothelial cells). Twenty-one breast cancer patient-derived xenografts as well as 2 transgenic mouse models (MMTV-PyMT and MMTV-ErbB2), and their respective allografts, were studied.Analysis of human and murine tumors showed a strong heterogeneity between tumors regarding infiltrating stroma-cells, with a high proportion of macrophages. A significant amount of polymorphonuclear leukocytes was also detected in PDXs, indicating a local inflammation in these models. The phenotypic analysis of macrophages showed a variable expression of M1 and M2 markers in PDXs. Macrophages infiltrating transgenic mouse tumors, spontaneous or allografted, were mainly M1. Transcriptomic analyses of sorted macrophages, allowed us to validate previous results but also highlighted major differences in the expression of numerous genes implicated in various pathways as tumor growth, invasion and metastasis.Finally, this study highlighted the impact of tumor cells on their surrounding stroma. Indeed, we demonstrate that cancer cells are able to attract a specific panel of stromal cells and activate them in a specific way
Lin, Shyr-Yeu 1962. "Genetically engineered mouse models for the study of follistatin biology." Monash University, Institute of Reproduction and Development, 2003. http://arrow.monash.edu.au/hdl/1959.1/5739.
Full textLambert, Laurens J. (Laurens Johannes). "Development and characterization of immunogenic genetically engineered mouse models of pancreatic cancer." Thesis, Massachusetts Institute of Technology, 2020. https://hdl.handle.net/1721.1/129020.
Full textCataloged from student-submitted PDF of thesis. Vita. Page 191 blank.
Includes bibliographical references.
Insights into mechanisms of immune escape have fueled the clinical success of immunotherapy in many cancers. However, pancreatic cancer has remained largely refractory to checkpoint immunotherapy. To uncover mechanisms of immune escape, we have characterized two preclinical models of immunogenic pancreatic ductal adenocarcinoma (PDAC). In order to dissect the endogenous antigen-specific T cell response in PDAC, lentivirus encoding the Cre recombinase and a tumor specific antigen (SIINFEKL, OVA[subscript 257-264]) was delivered to Kras[superscript LSL-G12D/+]; Trp[superscript 53flox/flox] (KP) mice. We demonstrate that KP tumors show distinct antigenic outcomes: a subset of PDAC tumors undergoes clearance or editing by a robust antigen-specific CD8+ T cell response, while a fraction undergo immune escape. Subsequently, we have developed an immunogenic pancreatic tumor organoid orthotopic transplant model.
In this model, immunogenic pancreatic tumors manifest divergent tumor phenotypes; 40% of tumor organoids do not form tumors ("non-progressors"), whereas 50% of organoids form aggressive tumors despite maintaining antigen expression and a demonstrable T cell response ("progressors"). Additionally, a subset (10%) of tumors show an intermediate phenotype, possibly reflective of an immune equilibrium state. We have further phenotypically and transcriptionally characterized the CD8+ T cell response to understand immune escape in this model. Our analyses reveal unexpected T cell heterogeneity, and acquisition of T cell dysfunctionality. Therapeutic combinatorial targeting of co-inhibitory receptors identified on dysfunctional antigen-specific CD8+ T cells led to dramatic regression of aggressive pancreatic tumors.
Finally, we demonstrate that human CD8+ T cells isolated from pancreatic tumors co-express co-inhibitory receptors, suggesting that T cell dysfunction may be operational in human disease. This is the first demonstration of immunoediting in an autochthonous and organoid-based model of pancreatic cancer. Further characterization of these preclinical model systems will enable rational design of novel clinical immunotherapeutic strategies for treatment of this devastating disease.
by Laurens J. Lambert.
Ph. D.
Ph.D. Massachusetts Institute of Technology, Department of Biology
Du, Page Michel Justin Porter. "Investigation of T cell-mediated immune surveillance against tumor-specific antigens in genetically engineered mouse models of cancer." Thesis, Massachusetts Institute of Technology, 2011. http://hdl.handle.net/1721.1/62620.
Full textThis electronic version was submitted by the student author. The certified thesis is available in the Institute Archives and Special Collections.
Cataloged from student-submitted PDF version of thesis. Vita.
Includes bibliographical references.
The association of tumor cells and lymphocytes has led to the hypothesis that our immune system actively inhibits the formation and progression of cancer, a phenomenon called tumor immune surveillance. T cells specific to mutant proteins have been identified in cancer patients and the recent success of cancer immunotherapies provides evidence that the immune system can fight this disease. Yet the frequent occurrence of malignant disease despite T cell recognition presents a significant medical problem. Only after we determine how tumors bypass the immune system can immunotherapeutic approaches be improved. To understand how tumors subvert immune responses, tumor transplantation or transgenic mice expressing tumor-associated antigens have been used to model cancer. To assess the role of anti-tumor T cells in models that more accurately reflect the human disease, I developed new systems to introduce exogenous antigens, to mimic neoantigens, into genetically engineered mouse models of lung cancer and sarcomas. Utilizing the mouse model of lung cancer, I show that endogenous T cells respond to and infiltrate lung tumors, delaying malignant progression. Despite continued antigen expression, T cell infiltration does not persist and tumors ultimately escape immune attack. Transplantation of cell lines derived from lung tumors that express these antigens or prophylactic vaccination against autochthonous tumors, however, results in rapid tumor eradication or selection of tumors that lose antigen expression. These results support clinical data that suggest a role for the immune system in cancer suppression rather than prevention. Tumor immune surveillance and immunoediting have largely been defined using carcinogen-driven models of sarcomagenesis. Using a genetically engineered model of sarcomagenesis, I show that immunoediting requires potent T cell antigens and that lymphocytes drive the evolution of less immunogenic tumors by selecting for antigen loss. Finally, immunotherapies have historically been ineffective in treating cancer patients. I show that vaccination against specific antigens expressed in mouse lung cancers leads to sustained anti-tumor T cell responses that eradicate recently initiated tumors. Vaccination also stimulates anti-tumor T cell responses in an antigen-independent fashion by enhancing the expansion and activity of T cells that recognize antigens only expressed in tumors.
by Michel Justin Porter Du Page.
Ph.D.
Schmidt, Leah Marie. "Investigating functions of tumor-infiltrating natural killer cells in genetically-engineered mouse models of non-small cell lung cancer." Thesis, Massachusetts Institute of Technology, 2016. http://hdl.handle.net/1721.1/104480.
Full textCataloged from PDF version of thesis. Vita.
Includes bibliographical references.
The immune system has long been hypothesized to play a role in restraining tumor growth, but compelling evidence for this role evaded scientists for the better part of a century. After many years of skepticism, the field of cancer immunology has recently undergone a major revolution. The success of modern immunotherapeutics has transformed the arenas of oncology and drug development. Large efforts are now focused on understanding the factors that dictate patient responses to immunotherapy, for the identification of possible points of intervention to expand the fraction of patients who benefit from therapy. The majority of approved immunotherapeutics directly target adaptive immune effectors. However, emerging evidence suggests that these treatments preferentially benefit patients with pre-existing immune responses against tumors, and patients who fail therapies often harbor tumors that are poorly infiltrated by adaptive immune cells. I have explored the role of an innate immune effector known for its capacity to kill tumor cells and its importance in stimulating and shaping adaptive immune responses, the natural killer (NK) cell. To this end, I developed a new system for assaying NK cell function in the context of established, autochthonous lung cancer, by engineering vectors for producing tumors with inducible NK cell activating ligands. Using this model, I have shown that NK cells in established tumors exhibit dysfunctional phenotypes, but their responses can be boosted by providing activating stimuli. Strikingly, stimulation of NK cells results in the recruitment of adaptive immune cells to tumors. By developing a next-generation model for inducing activating NK cell ligands in tumors engineered to express T cell antigens, I demonstrated that NK cell activation in immunogenic tumors results in effective immune responses that restrain tumor growth, highlighting the potential for cooperation between innate and adaptive arms of the immune system in anti-tumor immunity. Finally, I developed a novel immunotherapeutic molecule for stimulating NK cell responses against cancer cells. Bifunctional molecules are an emerging class of anti-cancer agents, designed to target immune effectors against tumors. I produced and performed initial functional testing on a bifunctional molecule that stimulates NK cell responses against tumors by 'decorating' the surface of cancer cells with activating NK cell ligands. I demonstrated that this bifunctional molecule induces NK cell cytotoxicity against tumor targets. Based on this work, we hypothesize that strategies for stimulating NK cells in tumors may enhance the efficacy of T cell-targeted therapies in the treatment of cancer.
by Leah Marie Schmidt.
Ph. D.
Lee, Tony. "Investigations into the role of nitric oxide in cardiovascular development and disease, insights gained from genetically engineered mouse models of human disease." Thesis, National Library of Canada = Bibliothèque nationale du Canada, 2000. http://www.collectionscanada.ca/obj/s4/f2/dsk1/tape4/PQDD_0019/MQ54141.pdf.
Full textGrüner, Barbara Maria [Verfasser], Angelika [Akademischer Betreuer] Schnieke, Michael [Akademischer Betreuer] Schemann, and Jens T. [Akademischer Betreuer] Siveke. "Molecular and proteomic analysis of signaling pathways in pancreatic ductal adenocarcinoma using genetically engineered mouse models / Barbara Maria Grüner. Gutachter: Angelika Schnieke ; Jens T. Siveke ; Michael Schemann. Betreuer: Michael Schemann." München : Universitätsbibliothek der TU München, 2012. http://d-nb.info/1031512365/34.
Full textSchönhuber, Nina Verfasser], Angelika [Akademischer Betreuer] [Schnieke, Dieter K. M. [Akademischer Betreuer] Saur, and Oliver [Akademischer Betreuer] Krämer. "Next-generation genetically engineered mouse models to study PI3K/3-phosphoinositide-dependent protein kinase 1 (Pdk1) signaling in pancreatic cancer / Nina Schönhuber. Betreuer: Dieter K. M. Saur. Gutachter: Oliver Krämer ; Dieter K. M. Saur ; Angelika Schnieke." München : Universitätsbibliothek der TU München, 2015. http://d-nb.info/1079654976/34.
Full textLin, Lu-Hsun, and 林呂勳. "Investigation of the Anti-Carcinogenesis potential of X Genetically Engineered Mouse (GEM) Model." Thesis, 2015. http://ndltd.ncl.edu.tw/handle/fr5cgq.
Full text國立陽明大學
生物藥學研究所
103
X gene is a DNA-binding protein mainly consists of an N-terminal transactivation domain and followed by a C-terminal region of DNA-binding domain. To understand the function of A mutation in X gene. We generate an X genetically engineered mice (X GEM). The lower cancer incidence phenotype in X GEM were be observed in our preliminary results. Interestingly, the expression microarray results were also identify important genes associated with carcinogenic. To study X GEM, which is relevant to the cancer incidence, we design an experiment to challenge cancer growth. Used chemical induction of liver cancer model to test the associated with carcinogenic in X GEM. The tumor size, number, phenotype and blood biochemistry were not significant differences between WT and X GEM mice, respectively. These results suggested that the X gene isn’t related to DEN induced HCC.
Books on the topic "Genetically-engineered mouse (GEM) models"
D, Hoit Brian, and Walsh Richard A. 1946-, eds. Cardiovascular physiology in the genetically engineered mouse. Norwell, MA: Kluwer Academic Publishers, 1998.
Find full textD, Hoit Brian, and Walsh Richard A. 1946-, eds. Cardiovascular physiology in the genetically engineered mouse. 2nd ed. Boston: Kluwer Academic Publishers, 2002.
Find full textCardiovascular Physiology in the Genetically Engineered Mouse (Developments in Cardiovascular Medicine). 2nd ed. Springer, 2001.
Find full textInvestigations into the role of nitric oxide in cardiovascular development and disease: Insights gained from genetically engineered mouse models of human disease. Ottawa: National Library of Canada, 2000.
Find full textBates, Gillian P., and Christian Landles. Preclinical Experimental Therapeutics. Oxford University Press, 2014. http://dx.doi.org/10.1093/med/9780199929146.003.0016.
Full textBook chapters on the topic "Genetically-engineered mouse (GEM) models"
Doetschman, Thomas, and L. Philip Sanford. "Overview of Designing Genetically Engineered Mouse (GEM) Models." In Genetically Engineered Mice for Cancer Research, 1–15. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_1.
Full textRosales, Cecilia, and Manel Esteller. "Epigenetic Mouse Models." In Genetically Engineered Mice for Cancer Research, 375–96. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_18.
Full textKucherlapati, Melanie, Ken Hung, Mari Kuraguchi, and Raju Kucherlapati. "Mouse Models for Colorectal Cancer." In Genetically Engineered Mice for Cancer Research, 309–29. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_15.
Full textPrescher, Jennifer A., and Christopher H. Contag. "Imaging Mouse Models of Human Cancer." In Genetically Engineered Mice for Cancer Research, 235–60. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_11.
Full textKadmiel, Mahita, Kimberly L. Fritz-Six, and Kathleen M. Caron. "Understanding RAMPs Through Genetically Engineered Mouse Models." In Advances in Experimental Medicine and Biology, 49–60. New York, NY: Springer US, 2012. http://dx.doi.org/10.1007/978-1-4614-2364-5_5.
Full textCarbajal, Eletha, and Eric C. Holland. "Mouse Models in Preclinical Drug Development: Applications to CNS Models." In Genetically Engineered Mice for Cancer Research, 549–67. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_26.
Full textLargaespada, David A. "Insertional Mutagenesis for Generating Mouse Models of Cancer." In Genetically Engineered Mice for Cancer Research, 57–82. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_4.
Full textHezel, Aram F., and Nabeel Bardeesy. "Genetically Engineered Mouse Models of Pancreatic Ductal Adenocarcinoma." In Tumor Models in Cancer Research, 377–95. Totowa, NJ: Humana Press, 2010. http://dx.doi.org/10.1007/978-1-60761-968-0_16.
Full textPhilip, Subha, and Shyam K. Sharan. "Using Recombineering Technology to Create Genetically Engineered Mouse Models." In Genetically Engineered Mice for Cancer Research, 37–56. New York, NY: Springer New York, 2011. http://dx.doi.org/10.1007/978-0-387-69805-2_3.
Full textSacca, Rosalba, Sandra J. Engle, Wenning Qin, Jeffrey L. Stock, and John D. McNeish. "Genetically Engineered Mouse Models in Drug Discovery Research." In Methods in Molecular Biology, 37–54. Totowa, NJ: Humana Press, 2009. http://dx.doi.org/10.1007/978-1-60761-058-8_3.
Full textConference papers on the topic "Genetically-engineered mouse (GEM) models"
Yamaguchi, Takashi, Sanae K. Ikehara, Hayao Nakanishi, and Yuzuru Ikehara. "Abstract 822: Genetically engineered mouse models of catastrophic pancreatic ductal adenocarcinoma." In Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA. American Association for Cancer Research, 2015. http://dx.doi.org/10.1158/1538-7445.am2015-822.
Full textWerr, LH, J. Ackermann, Y. Kahlert, J. Fischer, FT Wunderlich, and M. Fischer. "Investigating the role of activated telomerase in genetically engineered neuroblastoma mouse models." In 33. Jahrestagung der Kind-Philipp-Stiftung für pädiatr. onkolog. Forschung. © Georg Thieme Verlag KG, 2020. http://dx.doi.org/10.1055/s-0040-1709777.
Full textGupta, Aayush, Marija Trajkovic-Arsic, Irina Heid, Nicole Teichman, Evdokia Kalederis, Rickmer Braren, and Jens Siveke. "Abstract B141: Predictive value of genetically engineered endogenous mouse models in preclinical therapeutic studies." In Abstracts: AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; November 5-9, 2015; Boston, MA. American Association for Cancer Research, 2015. http://dx.doi.org/10.1158/1535-7163.targ-15-b141.
Full textVitucci, Mark, Byron Huff, Ryan E. Bash, Natalie O. Karpinich, Ralf S. Schmid, and C. Ryan Miller. "Abstract 4305: Dissecting the requirements for astrocytoma and invasion using genetically-engineered mouse models." In Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL. American Association for Cancer Research, 2012. http://dx.doi.org/10.1158/1538-7445.am2012-4305.
Full textChung, Wei-Jen, Jason Long, Jason Cheng, Chris Tran, Anwesha Dey, Anneleen Daemen, and Melissa Junttila. "Abstract 2987: Next generation sequencing analysis of genetically engineered mouse models of human cancers." In Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA. American Association for Cancer Research, 2015. http://dx.doi.org/10.1158/1538-7445.am2015-2987.
Full textScott, Clare L., Gwo Y. Ho, Elizabeth E. Lieschke, Olga Kondrashova, Ronny Drapkin, David Bowtell, and Matthew J. Wakefield. "Abstract 4798: Genetically engineered mouse models of proliferative C5 high grade serous ovarian cancer." In Proceedings: AACR Annual Meeting 2017; April 1-5, 2017; Washington, DC. American Association for Cancer Research, 2017. http://dx.doi.org/10.1158/1538-7445.am2017-4798.
Full textAbate-Shen, Cory, Alvaro Aytes Meneses, Carolyn W. Kinkade, Antonina Mitrofanova, Celine Lefebvre, Chee Wai Chua, Mireia Castillo-Martin, Edward Gelmann, Michael M. Shen, and Andrea Califano. "Abstract SY22-01: Interrogating gene expression programs from preclinical analyses of genetically engineered mouse models." In Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/1538-7445.am2011-sy22-01.
Full textChung, Wei-Jen, Anneleen Daemen, Jason Long, Jason Cheng, Chris Tran, Zora Modrusan, Oded Foreman, and Melissa Junttila. "Abstract 2686: The genomic landscape of Kras mutant genetically engineered mouse models of human cancers." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-2686.
Full textBurton, Liza J., Junghui Koo, Carol Tucker-Burden, Wei Zhou, Melissa Gilbert-Ross, Chunzi Huang, Gabriel Sica, and Adam Marcus. "Abstract 865: Isolation of circulating tumors cells from genetically engineered mouse models of lung adenocarcinoma." In Proceedings: AACR Annual Meeting 2019; March 29-April 3, 2019; Atlanta, GA. American Association for Cancer Research, 2019. http://dx.doi.org/10.1158/1538-7445.sabcs18-865.
Full textBurton, Liza J., Junghui Koo, Carol Tucker-Burden, Wei Zhou, Melissa Gilbert-Ross, Chunzi Huang, Gabriel Sica, and Adam Marcus. "Abstract 865: Isolation of circulating tumors cells from genetically engineered mouse models of lung adenocarcinoma." In Proceedings: AACR Annual Meeting 2019; March 29-April 3, 2019; Atlanta, GA. American Association for Cancer Research, 2019. http://dx.doi.org/10.1158/1538-7445.am2019-865.
Full text