Nikki Parker
Endocyte
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Nikki Parker.
International Journal of Cancer | 2007
Christopher P. Leamon; Joseph A. Reddy; Iontcho Radoslavov Vlahov; Elaine Westrick; Nikki Parker; Jeffrey S. Nicoson; Marilynn Vetzel
EC140 is a water soluble folate conjugate of desacetylvinblastine monohydrazide (DAVLBH), which is constructed with an endosome‐cleavable acyl hydrazone bond. This agent has proven to be active and specific against well established, subcutaneous folate receptor (FR)‐positive tumors in multiple animal models. Recent structure‐activity and optimization studies have yielded a disulfide bond‐containing counterpart to EC140, herein referred to as EC145. This new conjugate was found to retain high affinity for FR‐positive cells, and it produced specific, dose‐responsive activity in vitro. Comparative in vivo efficacy tests confirmed that, like EC140, EC145 displays activity against both syngeneic and xenograft tumor models. However, EC145 was found to be more active and better tolerated than EC140; hence, more durable complete responses were consistently observed in EC145‐treated tumor‐bearing animals. Furthermore, EC145 was not found to be active against a FR‐negative tumor model. Additional preclinical studies are therefore warranted to better understand EC145s breadth of activity against FR‐positive tumors.
Cancer Research | 2008
Christopher P. Leamon; Joseph A. Reddy; Marilynn Vetzel; Ryan Dorton; Elaine Westrick; Nikki Parker; Yu Wang; Iontcho Radoslavov Vlahov
The membrane-bound high-affinity folate receptor (FR) is highly expressed on a wide range of primary and metastatic human cancers, such as those originating in ovary, lung, breast, endometrium, kidney, and brain. Because folate-linked conjugates bind to and become internalized within FR-expressing cells (similar to that of free folic acid), we explored the possibility of using the folate ligand to target a potent, semisynthetic analogue of the microtubule inhibitor tubulysin B to FR-enriched tumors. When tested in vitro, a novel folate conjugate, herein referred to as EC0305, was found to specifically inhibit the growth of a panel of FR-positive cell lines (IC50 range, 1-10 nmol/L) in a dose-dependent manner, whereas cells lacking FR expression were unaffected. The potency of EC0305 was also confirmed against a human KB xenograft-nu/nu mouse cancer model. Here, a brief three times per week, 2-week regimen yielded remarkable antitumor activity (100% tumor-free animals) without causing significant weight loss or major organ tissue degeneration. In contrast, antitumor activity was completely abolished in EC0305-treated animals that were co-dosed with an excess of a nontoxic folate-containing analogue, thereby confirming that the antitumor effect of this agent was mediated by FRs. The advantage provided by folate conjugation was further proved by the untargeted free drug, which was found to be completely inactive at both tolerable and highly toxic dose levels. Collectively, these results show that this potent antiproliferative tubulysin compound can be specifically delivered to FR-positive tumors to provide substantial therapeutic benefit using well-tolerable dosing regimens.
Molecular Pharmaceutics | 2009
Joseph A. Reddy; Ryan Dorton; Alicia Dawson; Marilynn Vetzel; Nikki Parker; Jeffrey S. Nicoson; Elaine Westrick; Patrick J. Klein; Yu Wang; Iontcho Radoslavov Vlahov; Christopher P. Leamon
Herein we report on the potencies of 4 related folate-conjugated tubulysins constructed with either tubulysin B hydrazide (EC0305), tubulysin A hydrazide (EC0510), the N,O-acetal derivative of natural tubulysins (EC0317) or a tubulysin B ester (EC0302). Our results confirmed that EC0305 is the most favorable conjugate of the group due to its potent antitumor activity [100% cures at 1 micromol/kg, three times a week (TIW) for 2 weeks] and its favorably low toxicity profile. In contrast, the natural tubulysin B drug proved to be inactive against a human nasopharyngeal tumor model when administered at doses near to or greater than the maximum tolerated dose (MTD). When tested against more chemoresistant folate receptor expressing M109 and 4T1-cl2 tumors, EC0305 displayed superior antitumor activity over a previously disclosed folate conjugate of desacetylvinblastine monohydrazide (EC145). These studies demonstrate that EC0305 has significant antiproliferative activity against FR expressing tumors, including those which are generally more chemoresistant, and that EC0305 should be considered for development as a candidate for the treatment of advanced FR-expressing human cancers.
Journal of Pharmacology and Experimental Therapeutics | 2008
Christopher P. Leamon; Joseph A. Reddy; Ryan Dorton; Alicia Bloomfield; Kristen Emsweller; Nikki Parker; Elaine Westrick
Herein, we present a detailed analysis on the effects of feeding laboratory mice both high and low folic acid (folate)-containing diets as related to associated changes in serum and red blood cell (RBC) folate levels, tissue-derived folate receptor levels, and the ability of folate-drug conjugates to bind and effectuate activity against folate receptor (FR)-positive tumor xenografts. Our data show that serum and RBC folate concentrations sharply drop immediately after mice are switched to low folate diets; however, both parameters reach steady-state, “human-like” levels after 6 weeks. Interestingly, tissue-related folate binding capacities were also lowered during the dietary modulation period, whereas the net uptake of a radiolabeled folate conjugate was simultaneously increased 2.6- and 5-fold in FR-positive kidney and tumor tissue, respectively. Finally, the performances of several clinically and preclinically relevant folate-drug conjugates were evaluated against tumors in mice that were fed high or low folate diets. Except when administered at a dose level 6-fold less than that required to saturate endogenous FRs, no significant loss of antitumor activity was observed. From these findings, we conclude that lowering the dietary intake of folates in mice has little impact on the biological activity of repetitively dosed folate-targeted agents but that low folate diet regimens will reduce serum and RBC folate levels down to levels that more closely approximate the normal human ranges.
Journal of Pharmacology and Experimental Therapeutics | 2011
Christopher P. Leamon; Joseph A. Reddy; Patrick J. Klein; Iontcho Radoslavov Vlahov; Ryan Dorton; Alicia Bloomfield; Melissa Nelson; Elaine Westrick; Nikki Parker; Kristen Bruna; Marilynn Vetzel; Mark A Gehrke; Jeffrey S. Nicoson; Richard A. Messmann; Patricia LoRusso; Edward A. Sausville
During a phase I trial of EC145 (a folate-targeted vinca alkaloid conjugate), constipation was identified as the dose-limiting toxicity, probably from a nonfolate receptor-related liver clearance process capable of releasing unconjugated vinca alkaloid from EC145 and shuttling it to the bile. Here, we report on the selective placement of novel carbohydrate segments (1-amino-1-deoxy-glucitolyl-γ-glutamate) spaced in-between the folate and vinca alkaloid moieties of EC145, which yielded a new agent (EC0489) that is equipotent but less toxic than EC145. Whereas both compounds could cure tumor-bearing mice reproducibly, EC0489 differed from EC145 with i) a shorter elimination half-life, ii) approximately 70% decrease in bile clearance, iii) a 4-fold increase in urinary excretion, and iv) improved tolerability in rodents. This combination of improvements justified the clinical evaluation of EC0489 where currently administered dose levels have exceeded the maximal tolerated dose of EC145 by approximately 70%, thereby reflecting the translational benefits to this new approach.
Cancer Research | 2013
Deepika Dhawan; José A. Ramos-Vara; James F. Naughton; Liang Cheng; Philip S. Low; Ryan Rothenbuhler; Christopher P. Leamon; Nikki Parker; Patrick J. Klein; Iontcho Radoslavov Vlahov; Joseph A. Reddy; Michael O. Koch; Linda M. Murphy; Lindsey M. Fourez; Jane C. Stewart; Deborah W. Knapp
Folate receptors (FR) may be of use for targeted delivery of cytotoxic drugs in invasive urothelial carcinoma (iUC), for which improved therapy is needed. FR expression and function in iUC were explored and the antitumor activity and toxicity of a folate-targeted vinblastine conjugate were evaluated in dogs with naturally occurring iUC, an excellent model for human iUC. FR immunohistochemistry was carried out on iUC and normal human and dog bladder tissues together with nuclear scintigraphy in dogs to monitor iUC folate uptake. Dose escalation of a folate-targeted vinblastine compound, EC0905, was conducted in dogs with biopsy-confirmed, FR-positive iUC. FRs were detected by immunohistochemistry (PU17) in most primary iUC and many nodal and lung metastases from dogs, and scintigraphy confirmed folate uptake in both primary and metastatic lesions. The maximum tolerated dose of EC0905 in dogs was 0.25 mg/kg IV weekly, with neutropenia at higher doses. Tumor responses included partial remission (≥ 50% reduction in tumor volume) in five dogs and stable disease (<50% change in tumor volume) in four dogs. Immunoreactivity to PU17 was similar in humans (78% of primary iUC, 80% of nodal metastases). Less immunoreactivity to mab343 (22% of cases) occurred. FR-β was noted in 21% of human iUC cases. Our findings suggest folate-targeted therapy holds considerable promise for treating iUC, where FR-β may be important in addition to FR-α.
Molecular Cancer Therapeutics | 2006
Yingjuan Lu; Le Cun Xu; Nikki Parker; Elaine Westrick; Joseph A. Reddy; Marilynn Vetzel; Philip S. Low; Christopher P. Leamon
Folic acid (pteroylglutamic acid) represents a useful ligand for targeted cancer therapies because it binds to a common epithelial tumor antigen known as the folate receptor. We previously devised an immunotherapy strategy that uses a bispecific ligand, a folate-hapten (FITC) conjugate, to redirect endogenously induced anti-FITC antibodies to folate receptor–positive tumor cells following parenteral administration. Here, we present results from preclinical pharmacokinetic and tissue biodistribution studies using a radioactive folate-FITC conjugate and results from dose optimization studies done in tumor-bearing animals. Folate-FITC was found to be rapidly eliminated in non-immunized mice; however, in immunized hosts, folate-FITC was shown to form immune complexes with FITC-specific antibodies, the consequence of which was a ∼173-fold increase in drug exposure (i.e., area under the curve). Using a newly developed ELISA assay, the extent of circulating anti-FITC antibodies occupied by parenterally given folate-FITC was determined to be proportional to the given dose. Furthermore, high doses of folate-FITC were found to promote the cosaturation of tumor cell surface folate receptors and circulating FITC-specific antibodies, blocking the immune recognition of tumor cells and thereby reducing antitumor activity. Nonetheless, by extending the duration of treatment and administering subsaturating doses of folate-FITC, enhanced antitumor response was observed in mice bearing established folate receptor–positive M109 tumors. Overall, results from the present study may help to guide clinicians through on-going clinical investigations of folate-targeted immunotherapy. [Mol Cancer Ther 2006;5(12):3258–67]
Molecular Medicine | 2015
Yingjuan Lu; Nikki Parker; Paul J. Kleindl; Vicky A. Cross; Kristin Wollak; Elaine Westrick; Torian W. Stinnette; Mark A Gehrke; Kevin Wang; Hari Krishna R. Santhapuram; Fei You; Spencer Hahn; Jeremy F. Vaughn; Patrick J. Klein; Iontcho Radoslavov Vlahov; Philip S. Low; Christopher P. Leamon
Folate receptor (FR)-β has been identified as a promising target for antimacrophage and antiinflammatory therapies. In the present study, we investigated EC0565, a folic acid-derivative of everolimus, as a FR-specific inhibitor of the mammalian target of rapamycin (mTOR). Because of its amphiphilic nature, EC0565 was first evaluated for water solubility, critical micelle formation, stability in culture and FR-binding specificity. Using FR-expressing macrophages, the effect of EC0565 on mTOR signaling and cellular proliferation was studied. The pharmacokinetics, metabolism and bioavailability of EC0565 were studied in normal rats. The in vivo activity of EC0565 was assessed in rats with adjuvant arthritis, a “macrophage-rich” model with close resemblance to rheumatoid arthritis. EC0565 forms micellar aggregates in physiological buffers and demonstrates good water solubility as well as strong multivalent FR-binding capacity. EC0565 inhibited mTOR signaling in rat macrophages at nanomolar concentrations and induced G0/G1 cell cycle arrest in serum-starved RAW264.7 cells. Subcutaneously administered EC0565 in rats displayed good bioavailability and a relatively long half-life (∼12 h). When given at 250 nmol/kg, EC0565 selectively inhibited proliferating cell nuclear antigen expression in thioglycollate-stimulated rat peritoneal cells. With limited dosing regimens, the antiarthritic activity of EC0565 was found superior to that of etanercept, everolimus and a nontargeted everolimus analog. The in vivo activity of EC0565 was also comparable to that of a folate-targeted aminopterin. Folate-targeted mTOR inhibition may be an effective way of suppressing activated macrophages in sites of inflammation, especially in nutrient-deprived conditions, such as in the arthritic joints. Further investigation and improvement upon the physical and biochemical properties of EC0565 are warranted.
American Journal of Physiology-renal Physiology | 2018
Kevin R. Kipp; Samantha L Kruger; Margaret F Schimmel; Nikki Parker; Jonathan M. Shillingford; Christopher P. Leamon; Thomas Weimbs
Autosomal-dominant polycystic kidney disease (ADPKD) is a very common genetic disease leading to renal failure. Numerous aberrantly regulated signaling pathways have been identified as promising molecular drug targets for ADPKD therapy. In rodent models, many small-molecule drugs against such targets have proven effective in reducing renal cyst growth. For example, mammalian target of rapamycin (mTOR) inhibition with rapamycin greatly ameliorates renal cystic disease in several rodent models. However, clinical trials with mTOR inhibitors were disappointing largely due to the intolerable extrarenal side effects during long-term treatment with these drugs. Most other potential drug targets in ADPKD are also widely expressed in extrarenal tissues, which makes it likely that untargeted therapies with small-molecule inhibitors against such targets will lead to systemic adverse effects during the necessary long-term treatment of years and decades in ADPKD patients. To overcome this problem, we previously demonstrated that folate-conjugated rapamycin (FC-rapa) targets polycystic kidneys due to the high expression of the folate receptor (FRα) and that treatment of a nonortholgous PKD mouse model leads to inhibition of renal cyst growth. Here we show, in a head-to-head comparison with unconjugated rapamycin, that FCrapa inhibits renal cyst growth, mTOR activation, cell cycling, and fibrosis in an orthologous Pkd1 mouse model. Both unconjugated rapamycin and FC-rapa are similarly effective on polycystic kidneys in this model. However, FC-rapa lacks the extrarenal effects of unconjugated rapamycin, in particular immunosuppressive effects. We conclude that folate-conjugation is a promising avenue for increasing the tissue specificity of small-molecule compounds to facilitate very long-term treatment in ADPKD.
Cancer Research | 2017
Yingjuan Lu; Nikki Parker; Haiyan Chu; Michael R. Pugh; Satish Rao; Patrick J. Klein; Michael F. Ritchie; Lonnie Myer; Jennifer Jaskowiak; Christopher P. Leamon
Triple negative breast cancer (TNBC) patients are insensitive to hormonal or anti-HER2 therapy and have a higher recurrence rate among all breast cancer subtypes. There is a lack of common therapeutic targets in TNBC due to its six distinct molecular characteristics. Recently, ~50% of TNBC cases were found to express the folate receptor alpha (FRα) on tumor cells. FRα is a GPI-anchored membrane glycoprotein capable of bringing folate-targeted small-molecule drug conjugates (SMDCs) inside the cell. EC1456 is a folic acid-tubulysin B hydrazide (TubBH) SMDC that specifically binds to the membrane FRα and is internalized by endocytosis. While encapsulated within the early endosome, EC1456 releases TubBH into the cytosol where it inhibits the polymerization of tubulin into microtubules, thus blocking spindle formation to arrest cells in metaphase which ultimately induces apoptosis. EC1456 is currently under Phase 1 clinical investigation in patients with common solid tumors [IND# 118,859]. The purpose of this study is to evaluate EC1456 activity in Champions TumorGraftTM TNBC patient-derived xenograft (PDX) models to help guide our drug development strategies. These PDX models were derived from patients who were treated with multiple lines of standard-of-care agents. A total of six low-passage, FR-positive TNBC models were tested against two different treatment regimens of EC1456 (once or twice a week for 2 weeks only). Plasma and tumor drug concentrations were quantified by LC-MS/MS using satellite study animals. The tumor-bearing animals were monitored for up to 60 days to assess both short-term (i.e. % TGI) and long-term (%PR, CR, TFS) anti-tumor responses. Using a stringent efficacy criteria (≥60% CR/TFS), 3 of the 6 TNBC models were found highly sensitive to EC1456 and 3 were found resistant. To identify potential gene signatures of EC1456 response, bioinformatics analysis was performed using existing RNA-seq data and compared across a broad panel of TumorGraftTM TNBC models, regardless of FR expression status. Specific biomarkers of interest were further analyzed by qRT-PCR using control tumors from the current study. Together, our analysis revealed potential resistance mechanisms associated with microtubule dynamics as well as a cancer cell’s ability to undergo apoptosis. Citation Format: Yingjuan Lu, Nikki L. Parker, Haiyan Chu, Michael R. Pugh, Satish I. Rao, Patrick J. Klein, Michael F. Ritchie, Lonnie D. Myer, Jennifer Jaskowiak, Christopher P. Leamon. Evaluation of anti-tumor efficacy of EC1456 in low-passage and pre-treated patient-derived xenograft models of triple-negative breast 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 2057. doi:10.1158/1538-7445.AM2017-2057