Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where e Li is active.

Publication


Featured researches published by e Li.


Journal of Clinical Oncology | 2011

Phase I Pharmacokinetic and Pharmacodynamic Study of the Multikinase Inhibitor Sorafenib in Combination With Clofarabine and Cytarabine in Pediatric Relapsed/Refractory Leukemia

Hiroto Inaba; Jeffrey E. Rubnitz; Elaine Coustan-Smith; Lie Li; Brian Furmanski; Gerard P. Mascara; Kenneth M. Heym; Robbin Christensen; Mihaela Onciu; Sheila A. Shurtleff; Stanley Pounds; Ching-Hon Pui; Raul C. Ribeiro; Dario Campana; Sharyn D. Baker

PURPOSE To assess the toxicity, pharmacokinetics, and pharmacodynamics of multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in children with relapsed/refractory leukemia. PATIENTS AND METHODS Twelve patients with acute leukemia (11 with acute myeloid leukemia [AML]) received sorafenib on days 1 to 7 and then concurrently with cytarabine (1 g/m(2)) and clofarabine (stratum one: 40 mg/m(2), n = 10; stratum two [recent transplantation or fungal infection]: 20 mg/m(2), n = 2) on days 8 to 12. Sorafenib was continued until day 28 if tolerated. Two sorafenib dose levels (200 mg/m(2) and 150 mg/m(2) twice daily) were planned. Sorafenib pharmacokinetic and pharmacodynamic studies were performed on days 7 and 8. RESULTS At sorafenib 200 mg/m(2), two of four patients in stratum one and one of two patients in stratum two had grade 3 hand-foot skin reaction and/or rash as dose-limiting toxicities (DLTs). No DLTs were observed in six patients in stratum one at sorafenib 150 mg/m(2). Sorafenib inhibited the phosphorylation of AKT, S6 ribosomal protein, and 4E-BP1 in leukemia cells. The rate of sorafenib conversion to its metabolite sorafenib N-oxide was high (mean, 33%; range, 17% to 69%). In vitro, the N-oxide potently inhibited FLT3-internal tandem duplication (ITD; binding constant, 70 nmol/L) and the viability of five AML cell lines. On day 8, sorafenib decreased blast percentages in 10 of 12 patients (median, 66%; range, 9% to 95%). After combination chemotherapy, six patients (three FLT3-ITD and three FLT3 wild-type AML) achieved complete remission, two (both FLT3-ITD AML) had complete remission with incomplete blood count recovery, and one (FLT3 wild-type AML) had partial remission. CONCLUSION Sorafenib in combination with clofarabine and cytarabine is tolerable and shows activity in relapsed/refractory pediatric AML.


Nature Genetics | 2016

NUDT15 polymorphisms alter thiopurine metabolism and hematopoietic toxicity

Takaya Moriyama; Rina Nishii; Virginia Perez-Andreu; Wenjian Yang; Federico Antillon Klussmann; Xujie Zhao; Ting-Nien Lin; Keito Hoshitsuki; Jacob Nersting; Kentaro Kihira; Ute Hofmann; Yoshihiro Komada; Motohiro Kato; Robert McCorkle; Lie Li; Katsuyoshi Koh; Cesar R. Najera; Shirley Kow-Yin Kham; Tomoya Isobe; Zhiwei Chen; Edwynn Kean-Hui Chiew; Deepa Bhojwani; Cynthia Jeffries; Yan Lu; Matthias Schwab; Hiroto Inaba; Ching-Hon Pui; Mary V. Relling; Atsushi Manabe; Hiroki Hori

Widely used as anticancer and immunosuppressive agents, thiopurines have narrow therapeutic indices owing to frequent toxicities, partly explained by TPMT genetic polymorphisms. Recent studies identified germline NUDT15 variation as another critical determinant of thiopurine intolerance, but the underlying molecular mechanisms and the clinical implications of this pharmacogenetic association remain unknown. In 270 children enrolled in clinical trials for acute lymphoblastic leukemia in Guatemala, Singapore and Japan, we identified four NUDT15 coding variants (p.Arg139Cys, p.Arg139His, p.Val18Ile and p.Val18_Val19insGlyVal) that resulted in 74.4–100% loss of nucleotide diphosphatase activity. Loss-of-function NUDT15 diplotypes were consistently associated with thiopurine intolerance across the three cohorts (P = 0.021, 2.1 × 10−5 and 0.0054, respectively; meta-analysis P = 4.45 × 10−8, allelic effect size = −11.5). Mechanistically, NUDT15 inactivated thiopurine metabolites and decreased thiopurine cytotoxicity in vitro, and patients with defective NUDT15 alleles showed excessive levels of thiopurine active metabolites and toxicity. Taken together, these results indicate that a comprehensive pharmacogenetic model integrating NUDT15 variants may inform personalized thiopurine therapy.


Clinical Cancer Research | 2013

Contribution of OATP1B1 and OATP1B3 to the Disposition of Sorafenib and Sorafenib-Glucuronide

Eric I. Zimmerman; Shuiying Hu; Justin L. Roberts; Alice A. Gibson; Shelley Orwick; Lie Li; Alex Sparreboom; Sharyn D. Baker

Purpose: Many tyrosine kinase inhibitors (TKI) undergo extensive hepatic metabolism, but mechanisms of their hepatocellular uptake remain poorly understood. We hypothesized that liver uptake of TKIs is mediated by the solute carriers OATP1B1 and OATP1B3. Experimental Design: Transport of crizotinib, dasatinib, gefitinib, imatinib, nilotinib, pazopanib, sorafenib, sunitinib, vandetanib, and vemurafenib was studied in vitro using artificial membranes (PAMPA) and HEK293 cell lines stably transfected with OATP1B1, OATP1B3, or the ortholog mouse transporter, Oatp1b2. Pharmacokinetic studies were conducted with Oatp1b2-knockout mice and humanized OATP1B1- or OATP1B3-transgenic mice. Results: All 10 TKIs were identified as substrates of OATP1B1, OATP1B3, or both. Transport of sorafenib was investigated further, as its diffusion was particularly low in the PAMPA assay (<4%) than other TKIs that were transported by both OATP1B1 and OATP1B3. Whereas Oatp1b2 deficiency in vivo had minimal influence on parent and active metabolite N-oxide drug exposure, plasma levels of the glucuronic acid metabolite of sorafenib (sorafenib-glucuronide) were increased more than 8-fold in Oatp1b2-knockout mice. This finding was unrelated to possible changes in intrinsic metabolic capacity for sorafenib-glucuronide formation in hepatic or intestinal microsomes ex vivo. Ensuing experiments revealed that sorafenib-glucuronide was itself a transported substrate of Oatp1b2 (17.5-fold vs. control), OATP1B1 (10.6-fold), and OATP1B3 (6.4-fold), and introduction of the human transporters in Oatp1b2-knockout mice provided partial restoration of function. Conclusions: These findings signify a unique role for OATP1B1 and OATP1B3 in the elimination of sorafenib-glucuronide and suggest a role for these transporters in the in vivo handling of glucuronic acid conjugates of drugs. Clin Cancer Res; 19(6); 1458–66. ©2013 AACR.


Cancer Cell | 2015

Efficacy of Retinoids in IKZF1-Mutated BCR-ABL1 Acute Lymphoblastic Leukemia

Michelle L. Churchman; Jonathan Low; Chunxu Qu; Elisabeth Paietta; Lawryn H. Kasper; Yunchao Chang; Debbie Payne-Turner; Mark J. Althoff; Guangchun Song; Shann Ching Chen; Jing Ma; Michael Rusch; Dan McGoldrick; Michael Edmonson; Pankaj Gupta; Yong Dong Wang; William Caufield; Burgess B. Freeman; Lie Li; John C. Panetta; Sharyn D. Baker; Yung-Li Yang; Kathryn G. Roberts; Kelly McCastlain; Ilaria Iacobucci; Jennifer L. Peters; Victoria E. Centonze; Faiyaz Notta; Stephanie M. Dobson; Sasan Zandi

Alterations of IKZF1, encoding the lymphoid transcription factor IKAROS, are a hallmark of high-risk acute lymphoblastic leukemia (ALL), however the role of IKZF1 alterations in ALL pathogenesis is poorly understood. Here, we show that in mouse models of BCR-ABL1 leukemia, Ikzf1 and Arf alterations synergistically promote the development of an aggressive lymphoid leukemia. Ikzf1 alterations result in acquisition of stem cell-like features, including self-renewal and increased bone marrow stromal adhesion. Retinoid receptor agonists reversed this phenotype, partly by inducing expression of IKZF1, resulting in abrogation of adhesion and self-renewal, cell cycle arrest, and attenuation of proliferation without direct cytotoxicity. Retinoids potentiated the activity of dasatinib in mouse and human BCR-ABL1 ALL, providing an additional therapeutic option in IKZF1-mutated ALL.


Clinical Pharmacology & Therapeutics | 2013

Conjunctive Therapy of Cisplatin With the OCT2 Inhibitor Cimetidine: Influence on Antitumor Efficacy and Systemic Clearance

Jason A. Sprowl; L. van Doorn; Shuiying Hu; L van Gerven; P. de Bruijn; Lie Li; Alice A. Gibson; Ron H.J. Mathijssen; Alex Sparreboom

The organic cation transporter 2 (OCT2) regulates uptake of cisplatin in proximal tubules, and inhibition of OCT2 protects against severe cisplatin‐induced nephrotoxicity. However, it remains uncertain whether potent OCT2 inhibitors, such as cimetidine, can influence the antitumor properties and/or disposition of cisplatin. Using an array of preclinical assays, we found that cimetidine had no effect on the uptake and cytotoxicity of cisplatin in ovarian cancer cells with high OCT2 mRNA levels (IGROV‐1 cells). Moreover, the antitumor efficacy of cisplatin in mice bearing luciferase‐tagged IGROV‐1 xenografts was unaffected by cimetidine (P = 0.39). Data obtained in 18 patients receiving cisplatin (100 mg/m2) in a randomized crossover fashion with or without cimetidine (800 mg × 2) revealed that cimetidine did not alter exposure to unbound cisplatin, a marker of antitumor efficacy (4.37 vs. 4.38 µg·h/ml; P = 0.86). These results support the future clinical exploration of OCT2 inhibitors as specific modifiers of cisplatin‐induced nephrotoxicity.


Journal of Chromatography B | 2010

Quantitation of sorafenib and its active metabolite sorafenib N-oxide in human plasma by liquid chromatography-tandem mass spectrometry.

Lie Li; Ming Zhao; Fariba Navid; Keith W. Pratz; B. Doug Smith; Michelle A. Rudek; Sharyn D. Baker

A simple and rapid method with high performance liquid chromatography/tandem mass spectrometry is described for the quantitation of the kinase inhibitor sorafenib and its active metabolite sorafenib N-oxide in human plasma. A protein precipitation extraction procedure was applied to 50 μL of plasma. Chromatographic separation of the two analytes, and the internal standard [(2)H(3)(13)C]-sorafenib, was achieved on a C(18) analytical column and isocratic flow at 0.3 mL/min for 4 min. Mean within-run and between-run precision for all analytes were <6.9% and accuracy was <5.3%. Calibration curves were linear over the concentration range of 50-10,000 ng/mL for sorafenib and 10-2500 ng/mL for sorafenib N-oxide. This method allows a specific, sensitive, and reliable determination of the kinase inhibitor sorafenib and its active metabolite sorafenib N-oxide in human plasma in a single analytical run.


Clinical Cancer Research | 2012

Ontogeny and Sorafenib Metabolism

Eric I. Zimmerman; Justin L. Roberts; Lie Li; David Finkelstein; Alice A. Gibson; Amarjit S. Chaudhry; Erin G. Schuetz; Jeffrey E. Rubnitz; Hiroto Inaba; Sharyn D. Baker

Purpose: To investigate the role of ontogeny in sorafenib metabolism to the equipotent active metabolite sorafenib N-oxide. Experimental Design: Steady-state pharmacokinetic studies of sorafenib and metabolites were conducted in 30 children and young adults (17 males; median age, 9.5 years) receiving sorafenib 150 mg/m2 or 200 mg/m2 twice daily. Sorafenib metabolism was evaluated in vitro at 10 μmol/L using a panel of purified human cytochrome P450 (CYP) enzymes. Sorafenib metabolism and CYP3A4 expression was evaluated in 52 human liver samples from donors of ≤20 years old. The drug–drug interaction potential between sorafenib and azole antifungal agents was evaluated in vitro and in vivo. Results: No age-related differences in sorafenib apparent oral clearance were observed. Mean sorafenib N-oxide metabolite ratio was 0.27 ± 0.14. In children of ≤10 years of age, boys had approximately 2-fold higher N-oxide ratios than girls (0.40 ± 0.15 vs. 0.22 ± 0.12, P = 0.026). Of the CYPs evaluated, sorafenib was exclusively metabolized to sorafenib N-oxide by CYP3A4. A trend for increased N-oxide formation in boys was observed in liver samples, which correlated with CYP3A4 mRNA expression. Posaconazole and voriconazole potently inhibited sorafenib N-oxide formation in vitro, and reduced sorafenib N-oxide formation in 3 children given sorafenib concurrent with azoles. Conclusion: We have identified several factors affecting interpatient variability in sorafenib metabolism to the active N-oxide metabolite including age, sex, and concurrent treatment with azole antifungals. This knowledge may provide important considerations for the clinical use of sorafenib in children and possibly other kinase inhibitors undergoing CYP3A4-mediated metabolism. Clin Cancer Res; 18(20); 5788–95. ©2012 AACR.


Clinical Cancer Research | 2013

Contribution of Abcc4-Mediated Gastric Transport to the Absorption and Efficacy of Dasatinib

Brian Furmanski; Shuiying Hu; Ken-ichi Fujita; Lie Li; Alice A. Gibson; Laura J. Janke; Richard T. Williams; John D. Schuetz; Alex Sparreboom; Sharyn D. Baker

Purpose: Several oral multikinase inhibitors are known to interact in vitro with the human ATP-binding cassette transporter ABCC4 (MRP4), but the in vivo relevance of this interaction remains poorly understood. We hypothesized that host ABCC4 activity may influence the pharmacokinetic profile of dasatinib and subsequently affect its antitumor properties. Experimental Design: Transport of dasatinib was studied in cells transfected with human ABCC4 or the ortholog mouse transporter, Abcc4. Pharmacokinetic studies were done in wild-type and Abcc4-null mice. The influence of Abcc4 deficiency on dasatinib efficacy was evaluated in a model of Ph+ acute lymphoblastic leukemia by injection of luciferase-positive, p185(BCR-ABL)-expressing Arf(−/−) pre-B cells. Results: Dasatinib accumulation was significantly changed in cells overexpressing ABCC4 or Abcc4 compared with control cells (P < 0.001). Deficiency of Abcc4 in vivo was associated with a 1.75-fold decrease in systemic exposure to oral dasatinib, but had no influence on the pharmacokinetics of intravenous dasatinib. Abcc4 was found to be highly expressed in the stomach, and dasatinib efflux from isolated mouse stomachs ex vivo was impaired by Abcc4 deficiency (P < 0.01), without any detectable changes in gastric pH. Abcc4-null mice receiving dasatinib had an increase in leukemic burden, based on bioluminescence imaging, and decreased overall survival compared with wild-type mice (P = 0.048). Conclusions: This study suggests that Abcc4 in the stomach facilitates the oral absorption of dasatinib, and it possibly plays a similar role for other orally administered substrates, such as acetylsalicylic acid. This phenomenon also provides a mechanistic explanation for the malabsorption of certain drugs following gastric resection. Clin Cancer Res; 19(16); 4359–70. ©2013 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Mitigation of acute kidney injury by cell-cycle inhibitors that suppress both CDK4/6 and OCT2 functions

Navjotsingh Pabla; Alice A. Gibson; Mike Buege; Su Sien Ong; Lie Li; Shuiying Hu; Guoqing Du; Jason A. Sprowl; Aksana Vasilyeva; Laura J. Janke; Eberhard Schlatter; Taosheng Chen; Giuliano Ciarimboli; Alex Sparreboom

Significance Acute kidney injury (AKI) is a common clinical condition caused by loss of kidney function. Lack of therapeutic options has contributed to high mortality rates in AKI patients. Drug-induced AKI, as observed during cisplatin-based anticancer therapy, is responsible for about 20% of renal failure cases. The initial injury triggers a proliferative response in renal tubular cells, which in the presence of cellular damage can further accelerate renal injury. Our study provides evidence that the small-molecule cell-cycle inhibitors palbociclib and LEE011 can prevent cisplatin-induced AKI by inhibiting two relevant targets: renal cell-cycle progression and organic cation transporter 2, a renal uptake transporter of cisplatin. The future development of cyclin-dependent kinase 4/6 inhibitors as renal protective agents could have significant clinical benefits. Acute kidney injury (AKI) is a potentially fatal syndrome characterized by a rapid decline in kidney function caused by ischemic or toxic injury to renal tubular cells. The widely used chemotherapy drug cisplatin accumulates preferentially in the renal tubular cells and is a frequent cause of drug-induced AKI. During the development of AKI the quiescent tubular cells reenter the cell cycle. Strategies that block cell-cycle progression ameliorate kidney injury, possibly by averting cell division in the presence of extensive DNA damage. However, the early signaling events that lead to cell-cycle activation during AKI are not known. In the current study, using mouse models of cisplatin nephrotoxicity, we show that the G1/S-regulating cyclin-dependent kinase 4/6 (CDK4/6) pathway is activated in parallel with renal cell-cycle entry but before the development of AKI. Targeted inhibition of CDK4/6 pathway by small-molecule inhibitors palbociclib (PD-0332991) and ribociclib (LEE011) resulted in inhibition of cell-cycle progression, amelioration of kidney injury, and improved overall survival. Of additional significance, these compounds were found to be potent inhibitors of organic cation transporter 2 (OCT2), which contributes to the cellular accumulation of cisplatin and subsequent kidney injury. The unique cell-cycle and OCT2-targeting activities of palbociclib and LEE011, combined with their potential for clinical translation, support their further exploration as therapeutic candidates for prevention of AKI.


Clinical Cancer Research | 2014

Cisplatin-Induced Renal Injury is Independently Mediated by OCT2 and p53

Jason A. Sprowl; Cynthia S. Lancaster; Navjotsingh Pabla; Edwin Hermann; Ashley M. Kosloske; Alice A. Gibson; Lie Li; Dorothea Zeeh; Eberhard Schlatter; Laura J. Janke; Giuliano Ciarimboli; Alex Sparreboom

Purpose: Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2−/−mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2−/−mice, we hypothesized that alternate pathways are involved in the observed injury. Experimental Design: Studies were done in wild-type, Oct1/2−/−, or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays. Results: KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10−11) and Oct1/2−/−mice (P = 1.92 × 10−8). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2−/−mice (p53−/−/Oct1/2−/−) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53−/−/Oct1/2−/−mice. Conclusions: These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized. Clin Cancer Res; 20(15); 4026–35. ©2014 AACR.

Collaboration


Dive into the e Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alice A. Gibson

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Shuiying Hu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Navjotsingh Pabla

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Jason A. Sprowl

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Aksana Vasilyeva

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jeffrey E. Rubnitz

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Laura J. Janke

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge