1
|
Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, Duris C, North PE, Dalton S, Duncan SA. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology 2010; 51:297-305. [PMID: 19998274 PMCID: PMC2946078 DOI: 10.1002/hep.23354] [Citation(s) in RCA: 934] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED There exists a worldwide shortage of donor livers available for orthotropic liver transplantation and hepatocyte transplantation therapies. In addition to their therapeutic potential, primary human hepatocytes facilitate the study of molecular and genetic aspects of human hepatic disease and development and provide a platform for drug toxicity screens and identification of novel pharmaceuticals with potential to treat a wide array of metabolic diseases. The demand for human hepatocytes, therefore, heavily outweighs their availability. As an alternative to using donor livers as a source of primary hepatocytes, we explored the possibility of generating patient-specific human hepatocytes from induced pluripotent stem (iPS) cells. CONCLUSION We demonstrate that mouse iPS cells retain full potential for fetal liver development and describe a procedure that facilitates the efficient generation of highly differentiated human hepatocyte-like cells from iPS cells that display key liver functions and can integrate into the hepatic parenchyma in vivo.
Collapse
|
research-article |
15 |
934 |
2
|
Si-Tayeb K, Noto FK, Sepac A, Sedlic F, Bosnjak ZJ, Lough JW, Duncan SA. Generation of human induced pluripotent stem cells by simple transient transfection of plasmid DNA encoding reprogramming factors. BMC DEVELOPMENTAL BIOLOGY 2010; 10:81. [PMID: 20682060 PMCID: PMC2923111 DOI: 10.1186/1471-213x-10-81] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 08/03/2010] [Indexed: 12/11/2022]
Abstract
Background The use of lentiviruses to reprogram human somatic cells into induced pluripotent stem (iPS) cells could limit their therapeutic usefulness due to the integration of viral DNA sequences into the genome of the recipient cell. Recent work has demonstrated that human iPS cells can be generated using episomal plasmids, excisable transposons, adeno or sendai viruses, mRNA, or recombinant proteins. While these approaches offer an advance, the protocols have some drawbacks. Commonly the procedures require either subcloning to identify human iPS cells that are free of exogenous DNA, a knowledge of virology and safe handling procedures, or a detailed understanding of protein biochemistry. Results Here we report a simple approach that facilitates the reprogramming of human somatic cells using standard techniques to transfect expression plasmids that encode OCT4, NANOG, SOX2, and LIN28 without the need for episomal stability or selection. The resulting human iPS cells are free of DNA integration, express pluripotent markers, and form teratomas in immunodeficient animals. These iPS cells were also able to undergo directed differentiation into hepatocyte-like and cardiac myocyte-like cells in culture. Conclusions Simple transient transfection of plasmid DNA encoding reprogramming factors is sufficient to generate human iPS cells from primary fibroblasts that are free of exogenous DNA integrations. This approach is highly accessible and could expand the use of iPS cells in the study of human disease and development.
Collapse
|
Research Support, Non-U.S. Gov't |
15 |
170 |
3
|
DeLaForest A, Nagaoka M, Si-Tayeb K, Noto FK, Konopka G, Battle MA, Duncan SA. HNF4A is essential for specification of hepatic progenitors from human pluripotent stem cells. Development 2011; 138:4143-53. [PMID: 21852396 DOI: 10.1242/dev.062547] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The availability of pluripotent stem cells offers the possibility of using such cells to model hepatic disease and development. With this in mind, we previously established a protocol that facilitates the differentiation of both human embryonic stem cells and induced pluripotent stem cells into cells that share many characteristics with hepatocytes. The use of highly defined culture conditions and the avoidance of feeder cells or embryoid bodies allowed synchronous and reproducible differentiation to occur. The differentiation towards a hepatocyte-like fate appeared to recapitulate many of the developmental stages normally associated with the formation of hepatocytes in vivo. In the current study, we addressed the feasibility of using human pluripotent stem cells to probe the molecular mechanisms underlying human hepatocyte differentiation. We demonstrate (1) that human embryonic stem cells express a number of mRNAs that characterize each stage in the differentiation process, (2) that gene expression can be efficiently depleted throughout the differentiation time course using shRNAs expressed from lentiviruses and (3) that the nuclear hormone receptor HNF4A is essential for specification of human hepatic progenitor cells by establishing the expression of the network of transcription factors that controls the onset of hepatocyte cell fate.
Collapse
|
Research Support, Non-U.S. Gov't |
14 |
161 |
4
|
Cayo MA, Cai J, DeLaForest A, Noto FK, Nagaoka M, Clark BS, Collery RF, Si-Tayeb K, Duncan SA. JD induced pluripotent stem cell-derived hepatocytes faithfully recapitulate the pathophysiology of familial hypercholesterolemia. Hepatology 2012; 56:2163-71. [PMID: 22653811 PMCID: PMC3900031 DOI: 10.1002/hep.25871] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 05/24/2012] [Indexed: 12/14/2022]
Abstract
UNLABELLED Elevated levels of low-density lipoprotein cholesterol (LDL-C) in plasma are a major contributor to cardiovascular disease, which is the leading cause of death worldwide. Genome-wide association studies (GWAS) have identified 95 loci that associate with control of lipid/cholesterol metabolism. Although GWAS results are highly provocative, direct analyses of the contribution of specific allelic variations in regulating LDL-C has been challenging due to the difficulty in accessing appropriate cells from affected patients. The primary cell type responsible for controlling cholesterol and lipid flux is the hepatocyte. Recently, we have shown that cells with hepatocyte characteristics can be generated from human induced pluripotent stem cells (iPSCs). This finding raises the possibility of using patient-specific iPSC-derived hepatocytes to study the functional contribution of GWAS loci in regulating lipid metabolism. To test the validity of this approach, we produced iPSCs from JD a patient with mutations in the low-density lipoprotein receptor (LDLR) gene that result in familial hypercholesterolemia (FH). We demonstrate that (1) hepatocytes can be efficiently generated from FH iPSCs; (2) in contrast to control cells, FH iPSC-derived hepatocytes are deficient in LDL-C uptake; (3) control but not FH iPSC-derived hepatocytes increase LDL uptake in response to lovastatin; and (4) FH iPSC-derived hepatocytes display a marked elevation in secretion of lipidated apolipoprotein B-100. CONCLUSION Cumulatively, these findings demonstrate that FH iPSC-derived hepatocytes recapitulate the complex pathophysiology of FH in culture. These results also establish that patient-specific iPSC-derived hepatocytes could be used to definitively determine the functional contribution of allelic variation in regulating lipid and cholesterol metabolism and could potentially provide a platform for the identification of novel treatments of cardiovascular disease. (HEPATOLOGY 2012).
Collapse
|
research-article |
13 |
110 |
5
|
Cayo MA, Mallanna SK, Di Furio F, Jing R, Tolliver LB, Bures M, Urick A, Noto FK, Pashos EE, Greseth MD, Czarnecki M, Traktman P, Yang W, Morrisey EE, Grompe M, Rader DJ, Duncan SA. A Drug Screen using Human iPSC-Derived Hepatocyte-like Cells Reveals Cardiac Glycosides as a Potential Treatment for Hypercholesterolemia. Cell Stem Cell 2017; 20:478-489.e5. [PMID: 28388428 DOI: 10.1016/j.stem.2017.01.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/22/2016] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
Efforts to identify pharmaceuticals to treat heritable metabolic liver diseases have been hampered by the lack of models. However, cells with hepatocyte characteristics can be produced from induced pluripotent stem cells (iPSCs). Here, we have used hepatocyte-like cells generated from homozygous familial hypercholesterolemia (hoFH) iPSCs to identify drugs that can potentially be repurposed to lower serum LDL-C. We found that cardiac glycosides reduce the production of apolipoprotein B (apoB) from human hepatocytes in culture and the serum of avatar mice harboring humanized livers. The drugs act by increasing the turnover of apoB protein. Analyses of patient medical records revealed that the treatment of patients with cardiac glycosides reduced serum LDL-C levels. These studies highlight the effectiveness of using iPSCs to screen for potential treatments for inborn errors of hepatic metabolism and suggest that cardiac glycosides could provide an approach for reducing hepatocyte production of apoB and treating hypercholesterolemia.
Collapse
|
Research Support, Non-U.S. Gov't |
8 |
80 |
6
|
Gundry RL, Riordon DR, Tarasova Y, Chuppa S, Bhattacharya S, Juhasz O, Wiedemeier O, Milanovich S, Noto FK, Tchernyshyov I, Raginski K, Bausch-Fluck D, Tae HJ, Marshall S, Duncan SA, Wollscheid B, Wersto RP, Rao S, Van Eyk JE, Boheler KR. A cell surfaceome map for immunophenotyping and sorting pluripotent stem cells. Mol Cell Proteomics 2012; 11:303-16. [PMID: 22493178 DOI: 10.1074/mcp.m112.018135] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Induction of a pluripotent state in somatic cells through nuclear reprogramming has ushered in a new era of regenerative medicine. Heterogeneity and varied differentiation potentials among induced pluripotent stem cell (iPSC) lines are, however, complicating factors that limit their usefulness for disease modeling, drug discovery, and patient therapies. Thus, there is an urgent need to develop nonmutagenic rapid throughput methods capable of distinguishing among putative iPSC lines of variable quality. To address this issue, we have applied a highly specific chemoproteomic targeting strategy for de novo discovery of cell surface N-glycoproteins to increase the knowledge-base of surface exposed proteins and accessible epitopes of pluripotent stem cells. We report the identification of 500 cell surface proteins on four embryonic stem cell and iPSCs lines and demonstrate the biological significance of this resource on mouse fibroblasts containing an oct4-GFP expression cassette that is active in reprogrammed cells. These results together with immunophenotyping, cell sorting, and functional analyses demonstrate that these newly identified surface marker panels are useful for isolating iPSCs from heterogeneous reprogrammed cultures and for isolating functionally distinct stem cell subpopulations.
Collapse
|
Research Support, Non-U.S. Gov't |
13 |
55 |
7
|
El-Maghrabi MR, Noto F, Wu N, Manes N. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase: suiting structure to need, in a family of tissue-specific enzymes. Curr Opin Clin Nutr Metab Care 2001; 4:411-8. [PMID: 11568503 DOI: 10.1097/00075197-200109000-00012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present review addresses recent advances in research into a family of bifunctional enzymes that are responsible for the twofold task of synthesizing and hydrolyzing fructose-2,6-bisphosphate (Fru-2,6-P2), which in turn regulates the rate of glycolysis in most cells. The structure of the synthetic kinase, conjoined at its carboxyl-terminus to the phosphatase, is very highly conserved throughout evolution and differentiation, with isotypic expression arising from highly variable amino-terminal and carboxyl-terminal regulatory domains. These domains, which frequently contain protein-kinase-catalyzed phosphorylation motifs, are responsible for the widely divergent kinetics observed in various tissues and species, and for the hormonal modulation that alters intracellular levels of Fru-2,6-P2. The present review discusses recent advances in relating structure to function, and the identification of new pathways of transcriptional regulation of this important family of regulatory enzymes.
Collapse
|
Review |
24 |
29 |
8
|
Bonilla-Correal S, Noto F, Garcia-Bonavila E, Rodríguez-Gil JE, Yeste M, Miro J. First evidence for the presence of aquaporins in stallion sperm. Reprod Domest Anim 2017; 52 Suppl 4:61-64. [DOI: 10.1111/rda.13059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
|
8 |
17 |
9
|
Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY. Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Xenografts. Mol Cancer Ther 2018; 17:2481-2489. [PMID: 30206106 DOI: 10.1158/1535-7163.mct-18-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/10/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.
Collapse
|
Research Support, Non-U.S. Gov't |
7 |
17 |
10
|
Kasputis T, Clough D, Noto F, Rychel K, Dye B, Shea LD. Microporous Polymer Scaffolds for the Transplantation of Embryonic Stem Cell Derived Pancreatic Progenitors to a Clinically Translatable Site for the Treatment of Type I Diabetes. ACS Biomater Sci Eng 2018; 4:1770-1778. [PMID: 30345348 PMCID: PMC6191190 DOI: 10.1021/acsbiomaterials.7b00912] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Type I diabetes mellitus, which affects an estimated 1.5 million Americans, is caused by autoimmune destruction of the pancreatic beta cells that results in the need for life-long insulin therapy. Allogeneic islet transplantation for the treatment of type I diabetes is a therapy in which donor islets are infused intrahepatically, which has led to the transient reversal of diabetes. However, therapeutic limitations of allogeneic transplantation, which include a shortage of donor islets, long-term immunosuppression, and high risk of tissue rejection, have led to the investigation of embryonic or induced pluripotent stem cells as an unlimited source of functional beta-cells. Herein, we investigate the use of microporous scaffolds for their ability to promote the engraftment of stem cell derived pancreatic progenitors and their maturation toward mono-hormonal insulin producing β-cells at a clinically translatable, extrahepatic site. Initial studies demonstrated that microporous scaffolds supported cell engraftment, and their maturation to become insulin positive; however, the number of insulin positive cells and the levels of C-peptide secretion were substantially lower than what was observed with progenitor cell transplantation into the kidney capsule. The scaffolds were subsequently modified to provide a sustained release of exendin-4, which has previously been employed to promote maturation of pancreatic progenitors in vitro and has been employed to promote engraftment of transplanted islets in the peritoneal fat. Transplantation of stem cell derived pancreatic progenitors on scaffolds releasing exendin-4 led to significantly increased C-peptide production compared to scaffolds without exendin-4, with C-peptide and blood glucose levels comparable to the kidney capsule transplantation cohort. Image analysis of insulin and glucagon producing cells indicated that monohormonal insulin producing cells were significantly greater compared to glucagon producing and polyhormonal cells in scaffolds releasing exendin-4, whereas a significantly decreased percentage of insulin-producing cells were present among hormone producing cells in scaffolds without exendin-4. Collectively, a microporous scaffold, capable of localized and sustained delivery of exendin-4, enhanced the maturation and function of pluripotent stem cell derived pancreatic progenitors that were transplanted to a clinically translatable site.
Collapse
|
research-article |
7 |
15 |
11
|
Noto FK, Determan MR, Cai J, Cayo MA, Mallanna SK, Duncan SA. Aneuploidy is permissive for hepatocyte-like cell differentiation from human induced pluripotent stem cells. BMC Res Notes 2014; 7:437. [PMID: 25002137 PMCID: PMC4105394 DOI: 10.1186/1756-0500-7-437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 06/30/2014] [Indexed: 12/25/2022] Open
Abstract
Background The characterization of induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) routinely includes analyses of chromosomal integrity. The belief is that pluripotent stem cells best suited to the generation of differentiated derivatives should display a euploid karyotype; although, this does not appear to have been formally tested. While aneuploidy is commonly associated with cell transformation, several types of somatic cells, including hepatocytes, are frequently aneuploid and variation in chromosomal content does not contribute to a transformed phenotype. This insight has led to the proposal that dynamic changes in the chromosomal environment may be important to establish genetic diversity within the hepatocyte population and such diversity may facilitate an adaptive response by the liver to various insults. Such a positive contribution of aneuploidy to liver function raises the possibility that, in contrast to existing dogma, aneuploid iPSCs may be capable of generating hepatocyte-like cells that display hepatic activities. Results We examined whether a human iPSC line that had multiple chromosomal aberrations was competent to differentiate into hepatocytes and found that loss of normal chromosomal content had little impact on the production of hepatocyte-like cells from iPSCs. Conclusions iPSCs that harbor an abnormal chromosomal content retain the capacity to generate hepatocyte–like cells with high efficiency.
Collapse
|
Research Support, Non-U.S. Gov't |
11 |
13 |
12
|
Carluccio A, Noto F, Parrillo S, Contri A, De Amicis I, Gloria A, Robbe D, Veronesi MC. Transrectal ultrasonographic evaluation of combined utero-placental thickness during the last half of pregnancy in Martina Franca donkeys. Theriogenology 2016; 86:2296-2301. [PMID: 27566849 DOI: 10.1016/j.theriogenology.2016.07.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/21/2016] [Accepted: 07/21/2016] [Indexed: 01/10/2023]
Abstract
In the recent years, the donkey population decreased dramatically so that many breeds are presently considered as endangered. In comparison to the horse, the donkey placenta still remains not completely studied. In the horse, one of the diagnostic tools useful to identify pregnant mares at risk of abortion or premature delivery, include the transrectal ultrasound examination of the uterus and its contents; and especially of the combined thickness of the uterus and of the placenta (CUPT). Since the CUPT was never investigated in donkeys, the present study was aimed to define the transrectal CUPT values during the last half of pregnancy in 20 Martina Franca jennies. Foalings times, foals characteristics and placental gross appearance, and measurements were also evaluated and values resulted always within normality. Differently to the mare, a continuous significant CUPT increase between the sixth to the 12 months of pregnancy, and a substantial increase from the ninth to the 12th month of pregnancy, was found. Although statistically not evaluable, the CUPT values recorded from three jennies with pregnancy loss did not show evidence of CUPT increases. In conclusion, normal CUPT values from the sixth to the 12th month of pregnancy in Martina Franca donkeys are provided, but further investigations are needed to define possible breed or body-size CUPT specific differences, as well as the CUPT values during pregnancy disturbances or placental abnormalities.
Collapse
|
Journal Article |
9 |
12 |
13
|
Noto FK, Sangodkar J, Adedeji BT, Moody S, McClain CB, Tong M, Ostertag E, Crawford J, Gao X, Hurst L, O’Connor CM, Hanson EN, Izadmehr S, Tohmé R, Narla J, LeSueur K, Bhattacharya K, Rupani A, Tayeh MK, Innis JW, Galsky MD, Evers BM, DiFeo A, Narla G, Jamling TY. The SRG rat, a Sprague-Dawley Rag2/Il2rg double-knockout validated for human tumor oncology studies. PLoS One 2020; 15:e0240169. [PMID: 33027304 PMCID: PMC7540894 DOI: 10.1371/journal.pone.0240169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
We have created the immunodeficient SRG rat, a Sprague-Dawley Rag2/Il2rg double knockout that lacks mature B cells, T cells, and circulating NK cells. This model has been tested and validated for use in oncology (SRG OncoRat®). The SRG rat demonstrates efficient tumor take rates and growth kinetics with different human cancer cell lines and PDXs. Although multiple immunodeficient rodent strains are available, some important human cancer cell lines exhibit poor tumor growth and high variability in those models. The VCaP prostate cancer model is one such cell line that engrafts unreliably and grows irregularly in existing models but displays over 90% engraftment rate in the SRG rat with uniform growth kinetics. Since rats can support much larger tumors than mice, the SRG rat is an attractive host for PDX establishment. Surgically resected NSCLC tissue from nine patients were implanted in SRG rats, seven of which engrafted and grew for an overall success rate of 78%. These developed into a large tumor volume, over 20,000 mm3 in the first passage, which would provide an ample source of tissue for characterization and/or subsequent passage into NSG mice for drug efficacy studies. Molecular characterization and histological analyses were performed for three PDX lines and showed high concordance between passages 1, 2 and 3 (P1, P2, P3), and the original patient sample. Our data suggest the SRG OncoRat is a valuable tool for establishing PDX banks and thus serves as an alternative to current PDX mouse models hindered by low engraftment rates, slow tumor growth kinetics, and multiple passages to develop adequate tissue banks.
Collapse
|
Research Support, N.I.H., Extramural |
5 |
4 |
14
|
Scardina GA, Carini F, Noto F, Messina P. Microcirculation in the healing of surgical wounds in the oral cavity. Int J Oral Maxillofac Surg 2012; 42:31-5. [PMID: 22867738 DOI: 10.1016/j.ijom.2012.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/24/2012] [Accepted: 06/12/2012] [Indexed: 01/28/2023]
Abstract
The aim of this research is to evaluate in vivo the characteristics of microcirculation after taking a biopsy sample from the oral mucosa. 20 patients were recruited to the study and all underwent an oral mucosa biopsy for the excision of benign neoformations. The modifications in the oral microcirculation were evaluated in vivo in correspondence to the surgical site through videocapillaroscopy at three different times: 30 min before the biopsy; 48 h after the biopsy; and 7 days after the biopsy. The statistical significance was checked with the Mann-Whitney U-test (P<0.05). The analysis of videocapillaroscopic patterns showed statistically significant variations relative to the capillary loop density; the diameter of the outgoing loop; and the length of the capillary loop. In conclusion, the study describes a simple and reproducible model for the study of wound healing from a microcirculatory point of view.
Collapse
|
Journal Article |
13 |
4 |
15
|
O’Connor CM, Taylor SE, Miller KM, Hurst L, Haanen TJ, Suhan TK, Zawacki KP, Noto FK, Trako J, Mohan A, Sangodkar J, Zamarin D, DiFeo A, Narla G. Targeting Ribonucleotide Reductase Induces Synthetic Lethality in PP2A-Deficient Uterine Serous Carcinoma. Cancer Res 2022; 82:721-733. [PMID: 34921012 PMCID: PMC8857033 DOI: 10.1158/0008-5472.can-21-1987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/14/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022]
Abstract
Uterine serous carcinoma (USC) is a highly aggressive endometrial cancer subtype with limited therapeutic options and a lack of targeted therapies. While mutations to PPP2R1A, which encodes the predominant protein phosphatase 2A (PP2A) scaffolding protein Aα, occur in 30% to 40% of USC cases, the clinical actionability of these mutations has not been studied. Using a high-throughput screening approach, we showed that mutations in Aα results in synthetic lethality following treatment with inhibitors of ribonucleotide reductase (RNR). In vivo, multiple models of Aα mutant uterine serous tumors were sensitive to clofarabine, an RNR inhibitor (RNRi). Aα-mutant cells displayed impaired checkpoint signaling upon RNRi treatment and subsequently accumulated more DNA damage than wild-type (WT) cells. Consistently, inhibition of PP2A activity using LB-100, a catalytic inhibitor, sensitized WT USC cells to RNRi. Analysis of The Cancer Genome Atlas data indicated that inactivation of PP2A, through loss of PP2A subunit expression, was prevalent in USC, with 88% of patients with USC harboring loss of at least one PP2A gene. In contrast, loss of PP2A subunit expression was rare in uterine endometrioid carcinomas. While RNRi are not routinely used for uterine cancers, a retrospective analysis of patients treated with gemcitabine as a second- or later-line therapy revealed a trend for improved outcomes in patients with USC treated with RNRi gemcitabine compared with patients with endometrioid histology. Overall, our data provide experimental evidence to support the use of ribonucleotide reductase inhibitors for the treatment of USC. SIGNIFICANCE A drug repurposing screen identifies synthetic lethal interactions in PP2A-deficient uterine serous carcinoma, providing potential therapeutic avenues for treating this deadly endometrial cancer.
Collapse
|
Research Support, N.I.H., Extramural |
3 |
2 |
16
|
DeLaForest A, Nagaoka M, Si-Tayeb K, Noto FK, Konopka G, Battle MA, Duncan SA. HNF4A is essential for specification of hepatic progenitors from human pluripotent stem cells. J Cell Sci 2011. [DOI: 10.1242/jcs.099614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
|
14 |
1 |
17
|
Begemann D, Johnston N, O’Callaghan M, Walton G, Steffey V, Noto F. Abstract A040: In vivo imaging of ovarian and non-small cell lung cancer models hosted in the Sprague-Dawley Rag2 null Il2rgamma null SRG rat (OncoRat®). Cancer Res 2023. [DOI: 10.1158/1538-7445.metastasis22-a040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Abstract
Human cancer xenografts are a vital tool for understanding tumor biology. While in vivo studies are traditionally done in immunocompromised mice, we have created the SRG OncoRat® that is an excellent host for human xenografts. In vivo luminescent imaging is routinely used in mouse models, and it is particularly useful for studying orthotopic xenografts or metastatic models. In this study, we validated the SRG rat as a model that can effectively be used for in vivo imaging of human cancers. For the current study, we assessed two tumor models. OV81.2-luc is a luciferase positive cell line that was generated from ascites collection from a grade IIIC serous ovarian carcinoma. Tumors were established by inoculating OV81.2-luc cells IP into female SRG rats and female NSG mice, then treated with vehicle or cisplatin for 28 days. Human non-small cell lung cancer line H358-luc is luciferase positive and metastasizes to lung when injected subcutaneously into the hind flank of SRG rats. Tumors were measured thrice weekly with calipers. For both studies, we performed weekly in vivo luciferase imaging using Spectral Instruments AMI-HT. Tumors established rapidly for both OV81.2-luc and H358-luc in both SRG rats and NSG mice, with luciferin positive signal starting one week post inoculation. In OV81.2-luc hosting animals, upon necropsy, tumors were found on multiple abdominal organs including the peritoneum, ovary, mesentery, intestines, kidneys, liver, and body cavity wall. Metastatic events outside the abdominal cavity were not evident in OV81.2-luc hosting animals. In H358-luc tumor bearing rats, a high metastatic tumor burden was found in the lungs. These data confirm that the SRG rat is an excellent host for studying human cancer when compared to commonly used immunodeficient mouse models. Data demonstrate that the SRG rat has a high utility for studies using both in vivo imaging, such as orthotopic tumor implantation, and studies on metastasis. As the most immunodeficient rat commercially available, the SRG rat retains the ability to establish human tumors while possessing size, physiology, and metabolism-based advantages when compared to mice.
Citation Format: Diane Begemann, Nicolas Johnston, Marissa O’Callaghan, Grace Walton, Valeriya Steffey, Fallon Noto. In vivo imaging of ovarian and non-small cell lung cancer models hosted in the Sprague-Dawley Rag2 null Il2rgamma null SRG rat (OncoRat®) [abstract]. In: Proceedings of the AACR Special Conference: Cancer Metastasis; 2022 Nov 14-17; Portland, OR. Philadelphia (PA): AACR; Cancer Res 2022;83(2 Suppl_2):Abstract nr A040.
Collapse
|
|
2 |
|
18
|
Begemann D, Adedeji BT, Steffey V, Moody S, Narla G, Noto F. Abstract 2949: Sprague-Dawley Rag2 null Il2rgamma null SRG rat (OncoRat®) has enhanced tumor microenvironment in human prostate cancer xenografts. Cancer Res 2021. [DOI: 10.1158/1538-7445.am2021-2949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Human tumor xenografts are a staple tool for understanding tumor biology, growth kinetics, and therapeutic efficacy. While these studies are most commonly done in immunocompromised mice, we have created a Sprague Dawley Rag2 null, Il2rgamma null SRGTM rat that is an excellent host for human xenografts (OncoRat®). Lacking B, T, and NK cells, the SRG rat readily supports the growth of multiple human cancer cell lines, including lines that do not engraft well or grow consistently in existing mouse models. The tumor microenvironment (TME) is a critical factor for supporting xenograft tumors, and the microenvironment of a human tumor grown in the rat has yet to be fully characterized. In this study, a collaborative effort between research institutions discovered that the tumor microenvironment in the SRG rat is more robust, involved, and more supportive of human tumor growth than in NSG mice. To characterize the aforementioned differences in rat and mouse TME, human prostate cancer cell lines LNCaP and VCaP were grown in NSG mice and SRG rats. Formalin fixed paraffin embedded sections were stained via immunohistochemistry (IHC) for both rat and mouse tumor microenvironment markers. Collagen marker CD29, endothelial cell marker CD31, macrophage marker CD45, smooth muscle actin, and stromal markers CD54 and vimentin were analyzed in both animal hosts. When applicable, staining was quantified via counting positive cells per high powered field of view. When VCaP and LNCaP xenograft tumors are hosted by SRG rats, the host TME is significantly more involved within the human tumor, and readily supports tumor growth. Comparing the same markers in SRG rat and NSG mouse hosts revealed a stark difference - the SRG rat TME is more prevalent than the mouse. Results show significantly increased stromal cells per high powered field in the SRG rat when compared to tumors of the same cell line grown in the NSG mouse. There is heavily increased endothelial and stromal cell infiltration from the host into the human tumor, and higher heterogeneity within the cell population in tumors hosted by SRG rats. These data show that there are more human tumor epithelial cell interactions within the TME of the SRG rat than in NSG mouse. Increased stromal involvement more accurately recapitulates a human TME and may help explain the better take rates and faster growth rates of xenografts in SRG rats versus NSG mice. It is well known that recapitulating the tumor cell population heterogeneity is a study limitation when using animal models. Utilization of the SRG rat TME has great value in nonclinical research by more accurately translating into human disease, while remaining in a readily available immunodeficient animal model (i.e., OncoRat).
Citation Format: Diane Begemann, Bisoye Towobola Adedeji, Valeriya Steffey, Sam Moody, Goutham Narla, Fallon Noto. Sprague-Dawley Rag2 null Il2rgamma null SRG rat (OncoRat®) has enhanced tumor microenvironment in human prostate cancer xenografts [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2949.
Collapse
|
|
4 |
|
19
|
Noto FK, Ravi K, Arey A, McClain C, Zhang W, Narla G, Crawford J, Yeshi T. Abstract B36: Novel immunodeficient rat models capable of supporting the growth of human tumor xenografts. Cancer Res 2018. [DOI: 10.1158/1538-7445.mousemodels17-b36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Mouse models of human cancer have paved the way for studying cancer biology and genomics and their effects on cancer growth kinetics, propensity for metastasis, and treatment response. In addition, human cancer xenografts provide the opportunity to study cancer cell interactions with host stroma and tumor morphology. A plethora of genetically immunodeficient mouse models exist with different immune phenotypes, resulting in significant variability in tumor take rates and growth kinetics for a wide range of human cancer cell lines and patient-derived xenografts (PDX). Inconsistent or poor growth in these immunodeficient models have made downstream analysis and drug efficacy testing difficult. As a result, a significant number of mice are needed for drug efficacy screening to achieve a cohort of animals with tumors of similar size with similar tumor growth kinetics for treatment. It is possible that these cell lines might grow more consistently in a different immunodeficient model, such as an immunodeficient rat.
Until recently, the only immunodeficient rat that existed was the nude (NIH-Foxn1rnu; RNU) rat. This rat lacks T cells, but maintains a normal repertoire of all other immune cells, including B and NK cells. As such, there are a limited number of human cancer cell lines that can survive in the nude rat. We have created a genetically modified rat with a functional mutation of the Rag2 gene (Sprague Dawley – Rag2 null; SDR), resulting in a loss of mature B and T cells. In addition, we have created a Rag2/Il2rg double knockout rat (Sprague Dawley – Rag2; Il2rg null; SRG) that lacks mature B cells, T cells, and has fewer NK cells than wild-type Sprague Dawley rats. We have shown that the SDR rat is permissive for solid tumor growth of the human acute lymphocytic leukemia REH cell line, human glioblastoma U87MG cell line, human non-small cell lung cancer H358 cell line, and cell lines derived from ovarian and endometrial PDX samples. In some cases, tumor growth kinetics are superior in the SDR rat compared with immunodeficient mouse models. We have demonstrated that the human prostate cancer cell line, VCaP, which has poor engraftment efficiency and growth kinetics in the mouse, grows well in the SRG rat. The SRG rat is currently being validated for growth kinetics of other human cancer cell lines and PDX tissues. In addition, we are developing several disseminated human leukemia models in the SRG rat and creating immune-humanized mice and rats to be used in conjunction with human tumor xenografts for immunotherapy efficacy studies.
Citation Format: Fallon K. Noto, Kamesh Ravi, Angela Arey, Christopher McClain, Wei Zhang, Goutham Narla, Jack Crawford, Tseten Yeshi. Novel immunodeficient rat models capable of supporting the growth of human tumor xenografts [abstract]. In: Proceedings of the AACR Special Conference: Advances in Modeling Cancer in Mice: Technology, Biology, and Beyond; 2017 Sep 24-27; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(10 Suppl):Abstract nr B36.
Collapse
|
|
7 |
|
20
|
Noto FK, Adedeji BT, Agarwal Y, Beatty C, Ho S, Das A, Salunke R, Bility M, Jamling TY. Abstract A007: A Rag2/Il2rg double-knockout rat supports engraftment of human immune system for immunotherapy-based cancer efficacy studies. Mol Cancer Ther 2019. [DOI: 10.1158/1535-7163.targ-19-a007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Immune humanized mice have been valuable in the development of novel cancer immunotherapies and have demonstrated stronger efficacy when combined with standard of care chemotherapy. An immune humanized rat could provide several advantages over the currently available humanized mouse models, including supporting the growth of larger tumors for serial fine needle biopsies to assess immune infiltration and serial blood draws for assessing human immune development and tumor biomarkers in real-time throughout an efficacy study. We developed a novel autologous human skin and immune cells-humanized rat model by co-engrafting full-thickness human-fetal skin and autologous fetal lymphoid organoids under the kidney capsule along with intravenous injection of autologous fetal-liver derived hematopoietic stem cells, thus termed, human skin-immune system humanized rat model (hSIS-humanized rat). hSIS-humanized rat support development of adult-like, full-thickness human skin and human lymphoid organoids along with human immune cells. Methicillin-resistant Staphylococcus aureus inoculation in the human skin results in infection and skin pathology, thus recapitulating clinical outcomes. This model will enable in vivo mechanistic studies for development and evaluation of novel therapeutics for skin infectious disease and may also provide a model for establishing skin grafts of patient-derived melanoma tumors to investigate melanoma metastasis and response to therapies. In addition, engrafting the rat with human lymphoid organs and human immune cells may provide a similar platform to the BLT mouse for immunotherapy studies. Finally, we have demonstrated humanization of the rat immune system using human PBMCs. Human CD45+, CD3+, and CD20+ cells can be found in the peripheral blood, spleen, and bone marrow of engrafted rats. These immune humanized rat models may be beneficial for evaluating immunotherapies in human cancer models, including assessment of immune cell infiltration through fine needle biopsies.
Citation Format: Fallon K Noto, Bisoye Towobola Adedeji, Yash Agarwal, Cole Beatty, Sara Ho, Antu Das, Rajeev Salunke, Moses Bility, Tseten Yeshi Jamling. A Rag2/Il2rg double-knockout rat supports engraftment of human immune system for immunotherapy-based cancer efficacy studies [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A007. doi:10.1158/1535-7163.TARG-19-A007
Collapse
|
|
6 |
|
21
|
Ponnusamy SU, Noto FK, Adedeji BT, He Y, Hwang DJ, Moody S, Thiyagarajan T, Jamling TY, Miller DD, Narayanan R. Abstract B037: Potential next-generation androgen receptor-targeted therapeutic for enzalutamide-resistant prostate cancer; In vivo characterization in immune-compromised SRG rats. Mol Cancer Ther 2019. [DOI: 10.1158/1535-7163.targ-19-b037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Current treatment options for advanced castration-resistant prostate cancers (CRPC) are effective for a brief period before becoming refractory. It is important to use relevant in vivo models for candidate selection in the development of next-generation mechanistically-distinct drugs to treat castration- and drug- resistant prostate cancers. Here, we describe a series of AR pan-antagonists (selective AR degraders (SARDs)) that degrade the AR and AR splice variants. SARDs inhibit the wild-type and LBD mutant ARs comparably and inhibit the in vitro proliferation and in vivo growth of enzalutamide-sensitive and resistant prostate cancer xenografts. Xenograft studies conducted in immune-compromised SRG rats (Sprague Dawley Rag2-/- Il2rg -/-; Hera Biolabs) with three lead SARDs demonstrated regression of enzalutamide-sensitive and -resistant VCaP tumors both in castrated and in intact rats. SRG rats provide the benefit of abundant blood samples for weekly evaluation of rising PSA, which also indicated that the SARDs inhibit the rising PSA, while enzalutamide was inactive in enzalutamide-resistant model. Drug metabolism and pharmacokinetic (DMPK) studies, also conducted with SRG and wild-type rats and in combination with efficacy, indicate that the molecules possess all the necessary drug-like properties. The molecules exhibit a broad safety margin with no cross-reactivity with G-Protein Coupled Receptor, kinase, and nuclear receptor family members. Collectively, the SARDs exhibit the properties necessary for a next-generation prostate cancer drug and that use of SRG rats facilitated thorough characterization of their in vivo properties.
Citation Format: Suriyan Udhayasuriyan Ponnusamy, Fallon K Noto, Bisoye Towobola Adedeji, Yali He, Dong-Jin Hwang, Sam Moody, Thirumagal Thiyagarajan, Tseten Yeshi Jamling, Duane D Miller, Ramesh Narayanan. Potential next-generation androgen receptor-targeted therapeutic for enzalutamide-resistant prostate cancer; In vivo characterization in immune-compromised SRG rats [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B037. doi:10.1158/1535-7163.TARG-19-B037
Collapse
|
|
6 |
|
22
|
Noto FK, Arey A, McClain C, Zhang W, Yeshi T. Abstract 807: A novel immunodeficient rat for modeling human cancer. Cancer Res 2017. [DOI: 10.1158/1538-7445.am2017-807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Animal models of human cancer offer the potential to study human tumor growth kinetics, genetic variance among human cancers, and provide in vivo platforms for drug efficacy testing. In particular, immunodeficient mouse models have been invaluable in modeling a wide range of human cancers. However, some cancer lines don’t grow well in the available mouse models or show variability in growth kinetics from mouse to mouse, making drug efficacy studies difficult due to differences in tumor size at the onset of treatment. These challenges are also seen in patient derived xenograft (PDX) models, in addition to long timeframes to obtain sufficient mice with PDX tissue growth for drug efficacy studies. Mice are also limited in tumor growth potential with regard to humane endpoints and small size also limits the volume of blood that can be collected for analysis. An immunodeficient rat model could provide a solution to some of these issues. A rat model would allow for larger tumor size, easier surgical manipulation, and greater volume of tissue and blood sampling for downstream analysis. In addition, large tumors from rats could be serially transplanted into mice for drug efficacy testing and could provide a large number of transplanted mice in a shorter period of time compared with serially transplanting from mouse to mouse.
We have created an immunodeficient rat model with a functional deletion of the Rag2 gene. This knockout, created using spermatogonial stem cells, lacks mature B and T cells. To assess the capability of the Rag2 knockout rat to accept human xenografts, we transplanted 2 commercially available human cancer cell lines into our animals. The human REH acute lymphocytic leukemia cell line was transplanted via intravenous injection and the human glioblastoma cell line U87MG was transplanted subcutaneously. Both cell lines survived in the Rag2 knockout rat and resulted in the growth of tumors comprised of human cells. Studies are underway to characterize the Rag2 knockout rat’s ability to grow other human cell lines, including those that do not grow well in mice, and PDX tissues.
Citation Format: Fallon K. Noto, Angela Arey, Christopher McClain, Wei Zhang, Tseten Yeshi. A novel immunodeficient rat for modeling human cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 807. doi:10.1158/1538-7445.AM2017-807
Collapse
|
|
8 |
|
23
|
O'Connor CM, Zawacki KP, Durrant JR, Barrie K, Walton RG, King D, Onishi GH, Noto FK, Hinderman M, Begemann D, Branyan T, Panwell K, DiFeo A, Schlosser MJ, Narla G. The SRG RAT® supports human cell xenotransplantation through enhanced tumor microenvironment interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645250. [PMID: 40236141 PMCID: PMC11996424 DOI: 10.1101/2025.03.27.645250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The use of immunodeficient mice for human tumor engraftment is an essential model of human cancer, with uses ranging from basic science to translational research. However, low engraftment rates, slow growth, and smaller tumor volumes can be limitations. Previously, we reported a highly immunodeficient rat strain with the functional deletion of both the Rag2 and Il2rg genes on the Sprague-Dawley background ( SRG RAT ® ), which lacks B, T, and NK cells. Here, we subcutaneously engrafted two cell-derived xenograft (CDX) and seven patient-derived xenograft (PDX) models, including prostate, lung, ovarian, and uterine cancer models, into the SRG rat or NSG mouse models and tracked tumor growth. In all cases, the engraftment and tumor growth rates were better supported in the SRG rat compared to the NSG mouse. Interestingly, the SRG rat is not more immunocompromised than the NSG mouse, suggesting alternative mechanisms leading to the supportive growth in the SRG rat. Therefore, we explored potential differences in the tumor microenvironment (TME) between models grown in the two host animals. Lung PDX models grown in SRG rats showed enhanced formation of vasculature and stroma and were morphologically more consistent with the originating patient tumors. IHC analysis of the NCI-H660 CDX model showed differences in the tumors' stroma, vasculature, and macrophages when grown in the two host species. Single-cell spatial imaging of engrafted tumors showed upregulation of the human CXCL2 and TCIM in NCI-H660 tumors grown in the SRG rat versus the NSG mouse, both of which have been linked to poor prognosis in cancer. Combined, our data demonstrate that the SRG rat supports the growth of multiple human cancer types and displays enhanced tumor microenvironment interactions compared to NSG mice.
Collapse
|
Preprint |
1 |
|
24
|
Noto FK, Adedeji BT, Moody S, Brenzel C, Crawford J, Narla G, Jamling TY. Abstract 1059: A case study: OncoRat is a viable patient avatar for a NSCLC patient with a Y1248H Met activating mutation. Cancer Res 2019. [DOI: 10.1158/1538-7445.am2019-1059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Human cancer xenografts in rodents can provide predictive data on the success of candidate drugs in clinical trials and have been a pivotal tool in moving new drugs from the bench to the clinic. However, currently available immunodeficient mouse models have shown some limitation and variability in tumor take rates and growth kinetics in cancer cell lines. In addition, commercially available human cancer cell lines aren’t representative of the genomic and molecular diversity of cancers found in patients.
Patient Derived Xenograft (PDX), in which tumor tissue is transplanted directly into rodents after biopsy from the patient, better represents that molecular signature, heterogeneity, and pathology of the original tumor. Therefore, in vivo efficacy studies with PDX models could be highly predictive for treatment sensitivity. Despite the many advantages of PDXs for preclinical research, PDX mouse models are hindered by low engraftment rates and slow tumor growth kinetics. The loss of patient tumor heterogeneity and stromal cells as the PDX is passaged multiple times to generate sufficient tumor tissue to inoculate a cohort of animals for efficacy studies is also a disadvantage in the immunodeficient mouse models.
To address these limitations, we have introduced the OncoRat®; built on the SRGTM Platform, a Sprague-Dawley Rag2/Il2rg double knockout rat that lacks mature B cells, T cells, and circulating NK cells. We have demonstrated that the OncoRat has improved tumor take rate and growth kinetics for non-small cell lung cancer (NSCLC) PDXs. The NSLSC PDXs in the OncoRat have a much larger tumor volume, over 20,000 mm3 in the first passage (P0) in the rat, which provides an ample source of tissue for characterization and/or subsequent passage (P1) into OncoRat for drug efficacy studies. This leads to fewer animals used for study and faster timelines to drug efficacy data, resulting in a reduction in cost. In addition, we have used genomic analysis for guidance in planning in vivo efficacy studies. One of our NSCLC PDX models harbors a novel mutation in the MET pathway, suggesting this tumor would not be responsive to standard of care treatment. An efficacy study we performed in the OncoRat suggests that this particular tumor would respond well to Type II MET inhibitors, such as Cabonzantinib. This proof of concept study demonstrates that genomic and molecular analysis can provide insight into treatment outcomes and that PDX models in the OncoRat could serve as patient avatars for predicting treatment outcomes.
Citation Format: Fallon K. Noto, Bisoye Towobola Adedeji, Sam Moody, Chris Brenzel, Jack Crawford, Goutham Narla, Tseten Yeshi Jamling. A case study: OncoRat is a viable patient avatar for a NSCLC patient with a Y1248H Met activating mutation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1059.
Collapse
|
|
6 |
|
25
|
Gallina G, Cumbo V, Noto F, Messina P. [Recent advances in diagnosis of Sjogren's syndrome]. STOMATOLOGIA MEDITERRANEA : SM 1988; 8:321-32. [PMID: 3274169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this paper the authors review the classification and the clinical features of primary and secondary Sjogren's syndrome. Particular attention in focused on differential diagnosis, on clinical examination of the patients with chronic xerostomia and on its functional and pathological sequelae: rampant dental caries and oral candidiasis. Advanced in diagnostic procedures are described: salivary flow rate, salivary gland scintigraphy, parotid and submandibular scialography, Schirmer's test, Rose Bengal staining, minor salivary gland biopsy and serum immunological examinations. The relationships between scintigraphic and histologic result are discussed. The clinical features and the course of the disease provide further insight into the immunological pathogenesis of this syndrome.
Collapse
|
|
37 |
|