Fosciclopirox suppresses growth of high-grade urothelial cancer by targeting the γ-secretase complex

Scott J Weir, Prasad Dandawate, David Standing, Sangita Bhattacharyya, Prabhu Ramamoorthy, Parthasarathy Rangarajan, Robyn Wood, Amanda E Brinker, Benjamin L Woolbright, Mehmet Tanol, Tammy Ham, William McCulloch, Michael Dalton, Gregory A Reed, Michael J Baltezor, Roy A Jensen, John A Taylor 3rd, Shrikant Anant, Scott J Weir, Prasad Dandawate, David Standing, Sangita Bhattacharyya, Prabhu Ramamoorthy, Parthasarathy Rangarajan, Robyn Wood, Amanda E Brinker, Benjamin L Woolbright, Mehmet Tanol, Tammy Ham, William McCulloch, Michael Dalton, Gregory A Reed, Michael J Baltezor, Roy A Jensen, John A Taylor 3rd, Shrikant Anant

Abstract

Ciclopirox (CPX) is an FDA-approved topical antifungal agent that has demonstrated preclinical anticancer activity in a number of solid and hematologic malignancies. Its clinical utility as an oral anticancer agent, however, is limited by poor oral bioavailability and gastrointestinal toxicity. Fosciclopirox, the phosphoryloxymethyl ester of CPX (Ciclopirox Prodrug, CPX-POM), selectively delivers the active metabolite, CPX, to the entire urinary tract following parenteral administration. We characterized the activity of CPX-POM and its major metabolites in in vitro and in vivo preclinical models of high-grade urothelial cancer. CPX inhibited cell proliferation, clonogenicity and spheroid formation, and increased cell cycle arrest at S and G0/G1 phases. Mechanistically, CPX suppressed activation of Notch signaling. Molecular modeling and cellular thermal shift assays demonstrated CPX binding to γ-secretase complex proteins Presenilin 1 and Nicastrin, which are essential for Notch activation. To establish in vivo preclinical proof of principle, we tested fosciclopirox in the validated N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) mouse bladder cancer model. Once-daily intraperitoneal administration of CPX-POM for four weeks at doses of 235 mg/kg and 470 mg/kg significantly decreased bladder weight, a surrogate for tumor volume, and resulted in a migration to lower stage tumors in CPX-POM treated animals. This was coupled with a reduction in the proliferation index. Additionally, there was a reduction in Presenilin 1 and Hes-1 expression in the bladder tissues of CPX-POM treated animals. Following the completion of the first-in-human Phase 1 trial (NCT03348514), the pharmacologic activity of fosciclopirox is currently being characterized in a Phase 1 expansion cohort study of muscle-invasive bladder cancer patients scheduled for cystectomy (NCT04608045) as well as a Phase 2 trial of newly diagnosed and recurrent urothelial cancer patients scheduled for transurethral resection of bladder tumors (NCT04525131).

Conflict of interest statement

M.T. and S.J.W. are co-inventors of fosciclopirox composition of matter patents issued in the US, Europe, and Japan as well as a methods of use patents issued in the US. S.A. and S.J.W. are inventors on an issued patent in Europe, and a pending application in China. The exclusive, non-terminable rights to intellectual property generated by the inventors, as employees of the University, were licensed by the University of Kansas to CicloMed LLC in 2015. CicloMed is developing fosciclopirox for the treatment of bladder cancer. Kansas Life Sciences Development Company, a subsidiary of the University of Kansas Medical Center Research Institute, Inc. (which is a private, not-for-profit 501(c)(3) corporation established to promote and support medical research and faculty invention disclosures), possesses a financial interest in CicloMed.

Figures

Fig. 1. CPX inhibits proliferation and colony…
Fig. 1. CPX inhibits proliferation and colony formation in bladder cancer cells.
A Chemical structures of CPX and CPX-POM. B CPX inhibits proliferation of bladder cancer cell lines (T24, UM-UC-3, HTB-9, HTB-5, HT-1376, and RT-4). Cells were incubated with increasing concentration (0–40 μM) of CPX for up to 72 h. The treatment showed a significant dose and time-dependent decrease in cell proliferation when compared with untreated controls in the bladder cancer cell lines. C CPX inhibits colony formation. Cells were incubated with 0–20 μM CPX for 48 h and allowed to grow into colonies for 10 d. Incubation with CPX inhibits clonogenicity and number of colonies in bladder cancer cell lines (D).
Fig. 2. CPX induces cell cycle arrest…
Fig. 2. CPX induces cell cycle arrest in bladder cancer cell lines.
A Cell cycle analysis of CPX-treated cells. T24 and UM-UC-3 cells were treated with 4 µM concentrations of CPX, and examined by flow cytometry. CPX treatment-induced G0/G1 arrest in T24 cells and S-phase arrest in UM-UC-3 cells. The percent cells in each phase are presented graphically (B). C CPX downregulates cyclin D1 and B1 expression in T24 and UM-UC-3 cells as assessed by western blot. D Immunofluroscence analysis shows downregulation of cyclin D1 expression after CPX treatment in T24 cells.
Fig. 3. CPX induces apoptosis in bladder…
Fig. 3. CPX induces apoptosis in bladder cancer cell lines.
A T24 and UM-UC-3 cells were treated with CPX stained with Annexin V (FITC) and PI, and analyzed by flow cytometry. CPX treatment induced significant early and late apoptosis in T24 and UM-UC-3 cells. B The flow cytometric quantification of early and late apoptotic cells after CPX treatment over a period of 72 h in T24 and UM-UC-3 cells in Annexin-PI assay. C CPX induces apoptosis in T24 cells. T24 cells treated with CPX (4 µM) for 48 h were stained with Annexin V-FITC/PI solution and studied using immunofluorescence. CPX induced apoptosis in T24 cells. D Lysates from T24 and UM-UC-3 cells treated with CPX were studied by western blot for evaluating the effect on proteins involved in apoptosis. CPX treatment reduced the levels of antiapoptotic marker proteins Bcl2 and Bcl-XL. CPX treatment also increases the PARP cleavage compared to untreated controls. E CPX induces autophagy early which then followed by apoptosis. Lysates from T24 and UM-UC-3 cell treated with CPX (4 µM) for 24–48 h were analyzed by western blot. CPX treatment increased the expression of LC3B at 24 h and cleaved caspase-3 expression at 48 h. The data suggest that at early time points, CPX induces autophagy, which switches to apoptosis in T24 and UM-UC-3 cells.
Fig. 4. CPX inhibits bladder cancer spheroid…
Fig. 4. CPX inhibits bladder cancer spheroid growth and cell migration.
A, B CPX inhibits spheroid growth. CPX suppressed the size (A) and number (B) of spheroids of bladder cancer cell lines. Cells were grown in spheroid growth media in ultralow adherent plates and treated with CPX 2 µM and 4 µM concentrations for 5 days. CPX significantly decreased bladdosphere formation in the T24, UM-UC-3, HT-1376, HTB-9, and RT-4 cell lines. C CPX inhibits the expression of bladder cancer stem cell marker proteins. Lysates from T24 and UM-UC-3 cell lines after CPX treatment were subjected to western blot analysis. CPX treatment inhibited the expression of SOX9 and CD44 in both cell lines. D CPX inhibits migration of T24 and UM-UC-3 cells. Both cell lines were grown to 95–100% confluency and a scratch was made to study the cell movement over a period of 12 h. CPX treatment reduced cell migration. E CPX inhibited the percent migration in both cell lines. F CPX inhibits migration of T24 and UM-UC-3 cells. CPX treatment significantly reduced the migration of both cell lines through transwell cell culture inserts (Boyden’s chamber). G The percent migraition is represented in graphical format.
Fig. 5. CPX targets the Notch signaling…
Fig. 5. CPX targets the Notch signaling pathway, through inhibition of the γ-secretase complex.
A CPX inhibits mRNA expression of genes involved in Wnt, Hedgehog, and Notch cell signaling pathways. RNA extracted from T24 cells treated with and without CPX for 48 h was used to generate cDNA. The samples were analyzed using a Human stem cell RT2 profiler PCR array to identify signaling pathways that important for stem cell maintenance like Notch signaling and Wnt signaling. Expression of Hes5, Lfng, Myc, and Notch 1 mRNAs were significantly downregulated after CPX treatment. B CPX affects the expression of Notch receptors. Western blot analysis demonstrates that CPX treatment increased the expression of Notch 1 and suppressed the activation of Notch 2 and Notch 3, while CPX has little to no effect on Notch 4. C CPX suppresses γ-secretase pathway proteins. Western blot analysis of CPX-treated T24 and UM-UC-3 cell lysates showed CPX treatment reduced levels of Nicastrin, Presenilin 1, APH-1, and PEN-2. D CPX suppresses notch signaling pathway ligand and downstream proteins. CPX increased the expression of ligand Jagged 1 and inhibited the expression of downstream targets Hes1, cMYC, and Cyclin D1 in T24 and UM-UC-3 cells.
Fig. 6. CPX-POM treatment suppresses bladder tumorigenesis…
Fig. 6. CPX-POM treatment suppresses bladder tumorigenesis in vivo in the validated BBN mouse model of bladder cancer.
A Mean (±SD) plasma ciclopirox (CPX) concentration-time profiles following IV administration of 13.4 mg/kg CPX-POM and 18.1 mg/kg CPX-O to three C57BL/6 mice per serial blood collection time point per treatment group demonstrating rapid and complete metabolism of CPX-POM to its active metabolite, CPX. B Mean (±SD) plasma CPX concentration-time profiles following administration of 23.4 mg/kg IV and 117.5 mg/kg IP CPX-POM to three C57BL/6 mice per serial blood collection time point per treatment group demonstrating acceptable bioavailability of CPX following IP administration of CPX-POM. C Male C57BL/6 mice received 0.05% of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) in the drinking water for 16 weeks. Mice were treated with vehicle, 235 mg/kg CPX-POM (½MTD), or 470 mg/kg (MTD) CPX-POM IP once daily for 4 weeks (weeks 17–20). After 20 weeks, mice were euthanized, bladder was collected weighed, and subjected to further analysis. D CPX-POM treatment significantly reduced the bladder weight as compared to the vehicle-treated cohort at well-tolerated doses (p < 0.05). There were no statistically significant differences in bladder weights between the 235 mg/kg (½ MTD) and 470 mg/kg (MTD) CPX-POM treatment cohorts. E Immunohistochemistry analysis of bladder tumors obtained from CPX-POM-treated mice show a lower number of PCNA-positive nuclei compared to bladder tumors obtained from vehicle-treated mice. Bladder tumors obtained from CPX-POM treated mice also showed reduced expression of Notch 1, Presenilin 1, Hes1, and Nicastrin and increased levels of cleaved caspase-3 compared bladder tumors obtained from vehicle-treated mice.
Fig. 7. CPX targets γ-secretase complex proteins…
Fig. 7. CPX targets γ-secretase complex proteins Presenilin 1 and Nicastrin.
A Schematic representation showing the Notch signaling pathway. CPX targets γ-secretase target proteins Presenilin 1 and Nicastrin in bladder cancer cells. B mRNA expression of Presenilin 1 and Nicastrin in different cancer patients in the Cancer Genome Atlas database, extracted from Timer 2.0. (Bladder Urothelial Carcinoma [BLCA], Breast invasive carcinoma [BRCA], Cervical squamous cell carcinoma and endocervical adenocarcinoma [CESC], Colon Adenocarcinoma [COAD], Esophageal Squamous Cell Carcinoma [ESCA], Glioblastoma multiforme [GBM], Head and Neck squamous cell carcinoma [HNSC], Kidney renal clear cell carcinoma [KIRC], Liver hepatocellular carcinoma [LIHC], Lung adenocarcinoma [LUAD], Lung squamous cell carcinoma [LUSC], Pancreatic adenocarcinoma [PAAD], Prostate adenocarcinoma [PRAD], Rectum adenocarcinoma [READ], Stomach adenocarcinoma [STAD], Thyroid carcinoma [THEA], Uterine Corpus Endometrial Carcinoma [UCEC]). C Kaplan–Meier survival analysis showed that higher levels of Presenilin 1 or Nicastrin expression (p < 0.05) were significantly associated with poor overall survival in patients with bladder cancer. D The molecular docking analysis predicted the binding of CPX in the protein cavity of Presenilin 1 and Nicastrin. AutoDock Vina software program was used for molecular docking. E Cellular thermal shift assay (CETSA). T24 cell lysates were treated with CPX and subjected to differential temperature treatment for 3 mins and evaluated using western blot analyses. CPX protected Presenilin 1 and Nicastrin against thermal degradation, suggesting the active metabolite of CPX-POM interacts with these proteins. F Cumulative results from the densitometric evaluation of CETSA assay.

References

    1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J. Clin. 2021;71:7–33. doi: 10.3322/caac.21654.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J. Clin. 2019;69:7–34. doi: 10.3322/caac.21551.
    1. Botteman MF, Pashos CL, Redaelli A, Laskin B, Hauser R. The health economics of bladder cancer: a comprehensive review of the published literature. Pharmacoeconomics. 2003;21:1315–1330. doi: 10.1007/BF03262330.
    1. NCI. in Reports on Cancer: Annual Report to the Nation (NCI, 2018).
    1. Heney NM. Natural history of superficial bladder cancer. Prognostic features and long-term disease course. Urol. Clin. North Am. 1992;19:429–433. doi: 10.1016/S0094-0143(21)00411-0.
    1. Aldousari S, Kassouf W. Update on the management of non-muscle invasive bladder cancer. Can. Urol. Assoc. J. 2010;4:56–64. doi: 10.5489/cuaj.777.
    1. Millan-Rodriguez F, et al. Primary superficial bladder cancer risk groups according to progression, mortality and recurrence. J. Urol. 2000;164:680–684. doi: 10.1016/S0022-5347(05)67280-1.
    1. Grossman HB, et al. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N. Engl. J. Med. 2003;349:859–866. doi: 10.1056/NEJMoa022148.
    1. Gupta AK. Ciclopirox: an overview. Int. J. Dermatol. 2001;40:305–310. doi: 10.1046/j.1365-4362.2001.01156.x.
    1. Gupta AK, Sauder DN, Shear NH. Antifungal agents: an overview. Part I J. Am. Acad. Dermatol. 1994;30:677–698. doi: 10.1016/S0190-9622(08)81495-8.
    1. Eberhard Y, et al. Chelation of intracellular iron with the antifungal agent ciclopirox olamine induces cell death in leukemia and myeloma cells. Blood. 2009;114:3064–3073. doi: 10.1182/blood-2009-03-209965.
    1. Fujimura K, et al. A hypusine-eIF5A-PEAK1 switch regulates the pathogenesis of pancreatic cancer. Cancer Res. 2014;74:6671–6681. doi: 10.1158/0008-5472.CAN-14-1031.
    1. Goss KL, Gordon DJ. Gene expression signature based screening identifies ribonucleotide reductase as a candidate therapeutic target in Ewing sarcoma. Oncotarget. 2016;7:63003–63019. doi: 10.18632/oncotarget.11416.
    1. Koller CM, Kim Y, Schmidt-Wolf IG. Targeting renal cancer with a combination of WNT inhibitors and a bi-functional peptide. Anticancer Res. 2013;33:2435–2440.
    1. Liu Z, et al. The role of eukaryotic translation initiation factor 5A-1 (eIF5A-1) gene in HPV 16 E6 induces cell growth in human cervical squamous carcinoma cells. Biochem. Biophys. Res. Commun. 2018;504:6–12. doi: 10.1016/j.bbrc.2018.08.018.
    1. Memin E, et al. Blocking eIF5A modification in cervical cancer cells alters the expression of cancer-related genes and suppresses cell proliferation. Cancer Res. 2014;74:552–562. doi: 10.1158/0008-5472.CAN-13-0474.
    1. Mihailidou C, Papakotoulas P, Papavassiliou AG, Karamouzis MV. Superior efficacy of the antifungal agent ciclopirox olamine over gemcitabine in pancreatic cancer models. Oncotarget. 2018;9:10360–10374. doi: 10.18632/oncotarget.23164.
    1. Shen T, et al. Ciclopirox inhibits cancer cell proliferation by suppression of Cdc25A. Genes Cancer. 2017;8:505–516. doi: 10.18632/genesandcancer.135.
    1. Shen T, et al. Ciclopirox activates ATR-Chk1 signaling pathway leading to Cdc25A protein degradation. Genes Cancer. 2018;9:39–52. doi: 10.18632/genesandcancer.166.
    1. Sidarovich V, et al. Translational downregulation of HSP90 expression by iron chelators in neuroblastoma cells. Mol. Pharmacol. 2015;87:513–524. doi: 10.1124/mol.114.095729.
    1. Song S, et al. Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 2011;71:7628–7639. doi: 10.1158/0008-5472.CAN-11-2745.
    1. Wu J, et al. Antileukemia effect of ciclopirox olamine is mediated by downregulation of intracellular ferritin and inhibition beta-catenin-c-Myc signaling pathway in glucocorticoid resistant T-ALL cell lines. PLoS ONE. 2016;11:e0161509. doi: 10.1371/journal.pone.0161509.
    1. Yuan B, Ji W, Xia H, Li J. Combined analysis of gene expression and genome binding profiles identified potential therapeutic targets of ciclopirox in Ewing sarcoma. Mol. Med. Rep. 2018;17:4291–4298.
    1. Zhou H, et al. Ciclopirox olamine inhibits mTORC1 signaling by activation of AMPK. Biochem. Pharmacol. 2016;116:39–50. doi: 10.1016/j.bcp.2016.07.005.
    1. Zhou H, et al. The antitumor activity of the fungicide ciclopirox. Int. J. Cancer. 2010;127:2467–2477. doi: 10.1002/ijc.25255.
    1. Minden MD, et al. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies. Am. J. Hematol. 2014;89:363–368. doi: 10.1002/ajh.23640.
    1. Weir S. J. et al. Preclinical pharmacokinetics of fosciclopirox, a novel treatment for urothelial cancers in rats and dogs. J. Pharmacol. Exp. Ther.370, 148-159 (2019). doi: 10.1124/jpet.119.257972
    1. Stamatakos M, et al. Cell cyclins: triggering elements of cancer or not? World J. Surg. Oncol. 2010;8:111. doi: 10.1186/1477-7819-8-111.
    1. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu. Rev. Genet. 2009;43:67–93. doi: 10.1146/annurev-genet-102808-114910.
    1. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011;30:87. doi: 10.1186/1756-9966-30-87.
    1. Dandawate P, Padhye S, Ahmad A, Sarkar FH. Novel strategies targeting cancer stem cells through phytochemicals and their analogs. Drug Deliv. Transl. Res. 2013;3:165–182. doi: 10.1007/s13346-012-0079-x.
    1. Cooper CE, et al. The relationship of intracellular iron chelation to the inhibition and regeneration of human ribonucleotide reductase. J. Biol. Chem. 1996;271:20291–20299. doi: 10.1074/jbc.271.34.20291.
    1. Fryknäs M, et al. Iron chelators target both proliferating and quiescent cancer cells. Sci. Rep. 2016;6:38343–38343. doi: 10.1038/srep38343.
    1. Chestnut C, et al. Targeting major signaling pathways of bladder cancer with phytochemicals: a review. Nutr. Cancer. 2020;11:1–23. doi: 10.1080/01635581.2020.1856895.
    1. Hayashi T, et al. Not all NOTCH is created equal: the oncogenic role of NOTCH2 in bladder cancer and its implications for targeted therapy. Clin. Cancer Res. 2016;22:2981–2992. doi: 10.1158/1078-0432.CCR-15-2360.
    1. Yuan X, et al. Notch signaling: an emerging therapeutic target for cancer treatment. Cancer Lett. 2015;369:20–27. doi: 10.1016/j.canlet.2015.07.048.
    1. Patel MR, et al. Safety, dose tolerance, pharmacokinetics, and pharmacodynamics of fosciclopirox (CPX-POM) in patients with advanced solid tumors. J. Clin. Oncol. 2020;38:518–518. doi: 10.1200/JCO.2020.38.6_suppl.518.
    1. Hemming ML, Elias JE, Gygi SP, Selkoe DJ. Proteomic profiling of gamma-secretase substrates and mapping of substrate requirements. PLoS Biol. 2008;6:e257. doi: 10.1371/journal.pbio.0060257.
    1. Raman AE, Krishnan K, Maurya A, Sarkar N. In silico screening of drugs to find potential gamma-secretase inhibitors using pharmacophore modeling, QSAR and molecular docking studies. Comb. Chem. High. Throughput Screen. 2014;17:770–780. doi: 10.2174/1386207317666141019195448.
    1. Faltas BM, et al. Clonal evolution of chemotherapy-resistant urothelial carcinoma. Nat. Genet. 2016;48:1490. doi: 10.1038/ng.3692.
    1. Davarpanah NN, Yuno A, Trepel JB, Apolo AB. Immunotherapy: a new treatment paradigm in bladder cancer. Curr. Opin. Oncol. 2017;29:184–195. doi: 10.1097/CCO.0000000000000366.
    1. Suzman DL, et al. FDA Approval Summary: Atezolizumab or Pembrolizumab for the treatment of patients with advanced urothelial carcinoma ineligible for cisplatin-containing chemotherapy. Oncologist. 2019;24:563–569. doi: 10.1634/theoncologist.2018-0084.
    1. Maraver A, et al. NOTCH pathway inactivation promotes bladder cancer progression. J. Clin. Invest. 2015;125:824–830. doi: 10.1172/JCI78185.
    1. Rampias T, et al. A new tumor suppressor role for the Notch pathway in bladder cancer. Nat. Med. 2014;20:1199–1205. doi: 10.1038/nm.3678.
    1. Goriki A, et al. Unravelling disparate roles of NOTCH in bladder cancer. Nat. Rev. Urol. 2018;15:345–357. doi: 10.1038/s41585-018-0005-1.
    1. Zhang H, et al. Notch3 overexpression enhances progression and chemoresistance of urothelial carcinoma. Oncotarget. 2017;8:34362–34373. doi: 10.18632/oncotarget.16156.
    1. Hepburn AC, et al. Side population in human non-muscle invasive bladder cancer enriches for cancer stem cells that are maintained by MAPK signalling. PLoS ONE. 2012;7:e50690. doi: 10.1371/journal.pone.0050690.
    1. Dhareshwar SS, Stella VJ. Your prodrug releases formaldehyde: should you be concerned? No! J. Pharm. Sci. 2008;97:4184–4193. doi: 10.1002/jps.21319.
    1. DeGraff DJ, et al. Current preclinical models for the advancement of translational bladder cancer research. Mol. Cancer Ther. 2013;12:121–130. doi: 10.1158/1535-7163.MCT-12-0508.
    1. Li T, et al. TIMER: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 2017;77:e108–e110. doi: 10.1158/0008-5472.CAN-17-0307.
    1. Li T, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48:W509–W514. doi: 10.1093/nar/gkaa407.
    1. Landegren U. Measurement of cell numbers by means of the endogenous enzyme hexosaminidase. Applications to detection of lymphokines and cell surface antigens. J. Immunol. Methods. 1984;67:379–388. doi: 10.1016/0022-1759(84)90477-0.
    1. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput Chem. 2010;31:455–461.
    1. Xie T, et al. Crystal structure of the gamma-secretase component nicastrin. Proc. Natl Acad. Sci. USA. 2014;111:13349–13354. doi: 10.1073/pnas.1414837111.
    1. Pal D, et al. Targeting aberrant expression of Notch-1 in ALDH(+) cancer stem cells in breast cancer. Mol. Carcinog. 2017;56:1127–1136. doi: 10.1002/mc.22579.
    1. Alexander N, Woetzel N, Meiler J. bcl::Cluster: A method for clustering biological molecules coupled with visualization in the Pymol Molecular Graphics System. IEEE Int. Conf. Comput. Adv. Bio Med. Sci. 2011;2011:13–18.
    1. Jafari R, et al. The cellular thermal shift assay for evaluating drug target interactions in cells. Nat. Protoc. 2014;9:2100–2122. doi: 10.1038/nprot.2014.138.
    1. OECD. Test No. 425: Acute Oral Toxicity: Up-and-Down Procedure (2001).

Source: PubMed

3
Se inscrever