10 results on '"Daniel W. Pierce"'
Search Results
2. Phase I study of single-agent CC-292, a highly selective Bruton’s tyrosine kinase inhibitor, in relapsed/refractory chronic lymphocytic leukemia
- Author
-
Jennifer R. Brown, Wael A. Harb, Brian T. Hill, Janice Gabrilove, Jeff P. Sharman, Marshall T. Schreeder, Paul M. Barr, James M. Foran, Thomas P. Miller, Jan A. Burger, Kevin R. Kelly, Daruka Mahadevan, Shuo Ma, Yan Li, Daniel W. Pierce, Evelyn Barnett, Jeffrey Marine, Monika Miranda, Ada Azaryan, Xujie Yu, Pilar Nava-Parada, Jay Mei, and Thomas J. Kipps
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
3. Correction to: Quantitative Systems Pharmacology Modeling of Avadomide-Induced Neutropenia Enables Virtual Clinical Dose and Schedule Finding Studies
- Author
-
Roberto A. Abbiati, Michael Pourdehnad, Soraya Carrancio, Daniel W. Pierce, Shailaja Kasibhatla, Mark McConnell, Matthew W. B. Trotter, Remco Loos, Cristina C. Santini, and Alexander V. Ratushny
- Subjects
Neutropenia ,Dose-Response Relationship, Drug ,Neutrophils ,Pharmaceutical Science ,Correction ,Antineoplastic Agents ,Network Pharmacology ,Models, Biological ,Drug Administration Schedule ,Biological Variation, Population ,Humans ,Computer Simulation ,Piperidones ,Quinazolinones - Abstract
Avadomide is a cereblon E3 ligase modulator and a potent antitumor and immunomodulatory agent. Avadomide trials are challenged by neutropenia as a major adverse event and a dose-limiting toxicity. Intermittent dosing schedules supported by preclinical data provide a strategy to reduce frequency and severity of neutropenia; however, the identification of optimal dosing schedules remains a clinical challenge. Quantitative systems pharmacology (QSP) modeling offers opportunities for virtual screening of efficacy and toxicity levels produced by alternative dose and schedule regimens, thereby supporting decision-making in translational drug development. We formulated a QSP model to capture the mechanism of avadomide-induced neutropenia, which involves cereblon-mediated degradation of transcription factor Ikaros, resulting in a maturation block of the neutrophil lineage. The neutropenia model was integrated with avadomide-specific pharmacokinetic and pharmacodynamic models to capture dose-dependent effects. Additionally, we generated a disease-specific virtual patient population to represent the variability in patient characteristics and response to treatment observed for a diffuse large B-cell lymphoma trial cohort. Model utility was demonstrated by simulating the avadomide effect in the virtual population for various dosing schedules and determining the incidence of high-grade neutropenia, its duration, and the probability of recovery to low-grade neutropenia.
- Published
- 2022
4. A Platform for Rapid, Quantitative Assessment of Multiple Drug Combinations Simultaneously in Solid Tumors In Vivo.
- Author
-
Joyoti Dey, William S Kerwin, Marc O Grenley, Joseph R Casalini, Ilona Tretyak, Sally H Ditzler, Derek J Thirstrup, Jason P Frazier, Daniel W Pierce, Michael Carleton, and Richard A Klinghoffer
- Subjects
Medicine ,Science - Abstract
While advances in high-throughput screening have resulted in increased ability to identify synergistic anti-cancer drug combinations, validation of drug synergy in the in vivo setting and prioritization of combinations for clinical development remain low-throughput and resource intensive. Furthermore, there is currently no viable method for prospectively assessing drug synergy directly in human patients in order to potentially tailor therapies. To address these issues we have employed the previously described CIVO platform and developed a quantitative approach for investigating multiple combination hypotheses simultaneously in single living tumors. This platform provides a rapid, quantitative and cost effective approach to compare and prioritize drug combinations based on evidence of synergistic tumor cell killing in the live tumor context. Using a gemcitabine resistant model of pancreatic cancer, we efficiently investigated nine rationally selected Abraxane-based combinations employing only 19 xenografted mice. Among the drugs tested, the BCL2/BCLxL inhibitor ABT-263 was identified as the one agent that synergized with Abraxane® to enhance acute induction of localized apoptosis in this model of human pancreatic cancer. Importantly, results obtained with CIVO accurately predicted the outcome of systemic dosing studies in the same model where superior tumor regression induced by the Abraxane/ABT-263 combination was observed compared to that induced by either single agent. This supports expanded use of CIVO as an in vivo platform for expedited in vivo drug combination validation and sets the stage for performing toxicity-sparing drug combination studies directly in cancer patients with solid malignancies.
- Published
- 2016
- Full Text
- View/download PDF
5. ADAM12 is a circulating marker for stromal activation in pancreatic cancer and predicts response to chemotherapy
- Author
-
Peter Bailey, Maarten F. Bijlsma, Frederike Dijk, M.J. van de Vijver, Hanneke Wilmink, Andrew V. Biankin, R. Jiang, H.W.M. van Laarhoven, J. S. Li, Helene Damhofer, O.R.C. Busch, Cynthia Waasdorp, Daniel W. Pierce, Hemant M. Kocher, Marc G. Besselink, David K. Chang, Jan Paul Medema, Veronique L. Veenstra, L.B. van Rijssen, VU University medical center, Center of Experimental and Molecular Medicine, Graduate School, Surgery, AGEM - Digestive immunity, AGEM - Endocrinology, metabolism and nutrition, CCA - Imaging and biomarkers, AGEM - Re-generation and cancer of the digestive system, Pathology, Oncology, and Radiotherapy
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Stromal cell ,medicine.medical_treatment ,ADAM12 ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Stroma ,Internal medicine ,Pancreatic cancer ,Medicine ,Molecular Biology ,Chemotherapy ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Gemcitabine ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,business ,medicine.drug - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by abundant stroma that harbors tumor-promoting properties. No good biomarkers exist to monitor the effect of stromal targeting therapies or to predict response. We set out to identify such non-invasive markers for PDAC stroma and predict response to therapy. Gene expression datasets, co-culture experiments, xenografts, and patient samples were analyzed. Serum samples were measured from a cohort of 58 resected patients, and 87 metastatic or locally advanced PDAC patients. Baseline and follow-up levels were assessed in 372 additional metastatic PDAC patients who received nab-paclitaxel with gemcitabine (n = 184) or gemcitabine monotherapy (n = 188) in the phase III MPACT trial. Increased levels of ADAM12 were found in PDAC patients compared to healthy controls (p n = 157 and n = 38). High levels of ADAM12 significantly associated with poor outcome in resected PDAC (HR 2.07, p = 0.04). In the MPACT trial survival was significantly longer for patients who received nab-paclitaxel and had undetectable ADAM12 levels before treatment (OS 12.3 m vs 7.9 m p = 0.0046). Consistently undetectable or decreased ADAM12 levels during treatment significantly associated with longer survival as well (OS 14.4 m and 11.2 m, respectively vs 8.3, p = 0.0054). We conclude that ADAM12 is a blood-borne proxy for stromal activation, the levels of which have prognostic significance and correlate with treatment benefit.
- Published
- 2018
- Full Text
- View/download PDF
6. Phase I study of single-agent CC-292, a highly selective Bruton’s tyrosine kinase inhibitor, in relapsed/refractory chronic lymphocytic leukemia
- Author
-
Shuo Ma, James M. Foran, Thomas J. Kipps, Yan Li, Xujie Yu, Jeffrey Marine, Jeff P. Sharman, Jan A. Burger, Marshall T. Schreeder, Wael A. Harb, Paul M. Barr, Janice Gabrilove, Kevin R. Kelly, Jay Mei, Daruka Mahadevan, Thomas P. Miller, Evelyn Barnett, Pilar Nava-Parada, Daniel W. Pierce, Jennifer R. Brown, Monika Miranda, Ada Azaryan, and Brian T. Hill
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Population ,Neutropenia ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Bruton's tyrosine kinase ,education ,Online Only Articles ,Survival rate ,education.field_of_study ,biology ,business.industry ,Waldenstrom macroglobulinemia ,Hematology ,medicine.disease ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,Immunology ,biology.protein ,Mantle cell lymphoma ,business - Abstract
B-cell receptor (BCR) signaling plays a key role in the pathogenesis of B-cell malignancies, mediating the survival and proliferation of malignant B cells.1,2 Clinical studies have shown that Bruton’s tyrosine kinase (BTK) inhibitors are well tolerated, with promising clinical activity. Ibrutinib has shown 30-month progression-free survival (PFS) of 69% in relapsed chronic lymphocytic leukemia (CLL) patients,3–5 and has substantial activity in mantle cell lymphoma and activated B-cell-type diffuse large B-cell lymphoma.6,7 CC-292 is a highly selective oral small-molecule inhibitor that binds covalently and irreversibly to the same cysteine 481 in BTK as ibrutinib, inhibiting its signaling.8 We report here the results of a phase I study of CC-292 in patients with relapsed/refractory (R/R) CLL/small lymphocytic lymphoma (SLL), B-cell non-Hodgkin lymphoma (B-NHL), and Waldenstrom macroglobulinemia (WM). A total of 113 patients received continuous dosing with CC-292 in 28-day cycles at doses ranging from 125 mg to 1000 mg once daily, and 375 mg and 500 mg twice daily, continuing into dose-expansion cohorts of 750 mg once daily and a preliminary recommended phase II dose (RP2D)-expansion cohort of 500 mg twice daily. Four patients experienced dose-limiting toxicity (DLT) but only one in any treatment cohort. The most frequent grade 3–4 adverse events (AEs) were neutropenia (16%) and thrombocytopenia (8%). The most common non-hematologic treatment-emergent AEs (TEAEs) of any grade were diarrhea (68%) and fatigue (45%). Twice-daily administration of CC-292 was instituted to improve sustained BTK occupancy, and, in fact, did result in more than 90% BTK receptor occupancy at both the 4- and 24-h post-dose time points. Efficacy in the CLL/SLL population (n=84) showed that overall response rate (ORR) in patients receiving twice-daily dosing was 53%; an additional 10% had partial response with lymphocytosis (PR-L). CC-292 was, therefore, well tolerated and achieved high nodal and PR rates in relapsed CLL/SLL patients, but showed less durability than other BTK inhibitors.
- Published
- 2016
7. A Platform for Rapid, Quantitative Assessment of Multiple Drug Combinations Simultaneously in Solid Tumors In Vivo
- Author
-
Richard A. Klinghoffer, Sally Ditzler, Marc Grenley, Michael Carleton, Joyoti Dey, Ilona Tretyak, Jason Frazier, William S. Kerwin, Daniel W. Pierce, Derek Thirstrup, and Joseph Casalini
- Subjects
0301 basic medicine ,Oncology ,Cancer Treatment ,lcsh:Medicine ,Apoptosis ,Pharmacology ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine and Health Sciences ,Quantitative assessment ,Drug Interactions ,lcsh:Science ,media_common ,Sulfonamides ,Aniline Compounds ,Multidisciplinary ,Cell Death ,Pharmaceutics ,Drug Synergism ,Microinjection ,Synergy ,Cell Processes ,030220 oncology & carcinogenesis ,Research Article ,medicine.drug ,Drug ,medicine.medical_specialty ,Drug Research and Development ,Drug Administration ,media_common.quotation_subject ,Context (language use) ,Research and Analysis Methods ,Pancreatic Cancer ,03 medical and health sciences ,Drug Therapy ,In vivo ,Pancreatic cancer ,Internal medicine ,Gastrointestinal Tumors ,medicine ,Animals ,Molecular Biology Techniques ,Molecular Biology ,business.industry ,lcsh:R ,Biology and Life Sciences ,Cancers and Neoplasms ,Cancer ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,Pancreatic Neoplasms ,030104 developmental biology ,Drug Resistance, Neoplasm ,lcsh:Q ,Albumin-Bound Paclitaxel ,business - Abstract
While advances in high-throughput screening have resulted in increased ability to identify synergistic anti-cancer drug combinations, validation of drug synergy in the in vivo setting and prioritization of combinations for clinical development remain low-throughput and resource intensive. Furthermore, there is currently no viable method for prospectively assessing drug synergy directly in human patients in order to potentially tailor therapies. To address these issues we have employed the previously described CIVO platform and developed a quantitative approach for investigating multiple combination hypotheses simultaneously in single living tumors. This platform provides a rapid, quantitative and cost effective approach to compare and prioritize drug combinations based on evidence of synergistic tumor cell killing in the live tumor context. Using a gemcitabine resistant model of pancreatic cancer, we efficiently investigated nine rationally selected Abraxane-based combinations employing only 19 xenografted mice. Among the drugs tested, the BCL2/BCLxL inhibitor ABT-263 was identified as the one agent that synergized with Abraxane® to enhance acute induction of localized apoptosis in this model of human pancreatic cancer. Importantly, results obtained with CIVO accurately predicted the outcome of systemic dosing studies in the same model where superior tumor regression induced by the Abraxane/ABT-263 combination was observed compared to that induced by either single agent. This supports expanded use of CIVO as an in vivo platform for expedited in vivo drug combination validation and sets the stage for performing toxicity-sparing drug combination studies directly in cancer patients with solid malignancies.
- Published
- 2016
8. A Developmentally Regulated Kinesin-related Motor Protein from Dictyostelium discoideum
- Author
-
Ronald D. Vale, Eugenio L. de Hostos, Richard Sucgang, Gretchen McCaffrey, and Daniel W. Pierce
- Subjects
Population ,Molecular Sequence Data ,Motility ,Kinesins ,Microtubules ,Polymerase Chain Reaction ,Dictyostelium discoideum ,Article ,Motor protein ,Microtubule ,Null cell ,Animals ,Dictyostelium ,Amino Acid Sequence ,Cloning, Molecular ,education ,Molecular Biology ,Conserved Sequence ,education.field_of_study ,biology ,Sequence Homology, Amino Acid ,fungi ,Cell Polarity ,Gene Expression Regulation, Developmental ,Cell Biology ,biology.organism_classification ,Recombinant Proteins ,Cell biology ,Kinesin ,Microtubule-Associated Proteins ,Sequence Alignment - Abstract
The cellular slime mold Dictyostelium discoideum is an attractive system for studying the roles of microtubule-based motility in cell development and differentiation. In this work, we report the first molecular characterization of kinesin-related proteins (KRPs) in Dictyostelium. A PCR-based strategy was used to isolate DNA fragments encoding six KRPs, several of which are induced during the developmental program that is initiated by starvation. The complete sequence of one such developmentally regulated KRP (designated K7) was determined and found to be a novel member of the kinesin superfamily. The motor domain of K7 is most similar to that of conventional kinesin, but unlike conventional kinesin, K7 is not predicted to have an extensive α-helical coiled-coil domain. The nonmotor domain is unusual and is rich in Asn, Gln, and Thr residues; similar sequences are found in other developmentally regulated genes inDictyostelium. K7, expressed in Escherichia coli, supports plus end–directed microtubule motility in vitro at a speed of 0.14 μm/s, indicating that it is a bona fide motor protein. The K7 motor is found only in developing cells and reaches a peak level of expression between 12 and 16 h after starvation. By immunofluorescence microscopy, K7 localizes to a membranous perinuclear structure. To examine K7 function, we prepared a null cell line but found that these cells show no gross developmental abnormalities. However, when cultivated in the presence of wild-type cells, the K7-null cells are mostly absent from the prestalk zone of the slug. This result suggests that in a population composed largely of wild-type cells, the absence of the K7 motor protein interferes either with the ability of the cells to localize to the prestalk zone or to differentiate into prestalk cells.
- Published
- 1998
9. The Directional Preference of Kinesin Motors Is Specified by an Element outside of the Motor Catalytic Domain
- Author
-
Nora Hom-Booher, Daniel W. Pierce, Ronald D. Vale, Ryan B. Case, and Cynthia L. Hart
- Subjects
Polarity (physics) ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Kinesin 13 ,Kinesins ,macromolecular substances ,General Biochemistry, Genetics and Molecular Biology ,Domain (software engineering) ,Microtubule ,Escherichia coli ,Animals ,Drosophila Proteins ,Humans ,Directionality ,Amino Acid Sequence ,Adenosine Triphosphatases ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Processivity ,Protein Structure, Tertiary ,Tubulin ,Biochemistry ,Biophysics ,biology.protein ,Kinesin ,Drosophila - Abstract
Members of the kinesin superfamily share a similar motor catalytic domain yet move either toward the plus end (e.g., conventional kinesin) or the minus end (e.g., Ncd) of microtubules. The structural features that determine the polarity of movement have remained enigmatic. Here, we show that kinesin's catalytic domain (316 residues) in a dimeric construct (560 residues) can be replaced with the catalytic domain of Ncd and that the resultant motor moves in the kinesin direction. We also demonstrate that this chimera does not move processively over many tubulin subunits, which is similar to Ncd but differs from the highly processive motion of conventional kinesin. These findings reveal that the catalytic domain contributes to motor processivity but does not control the polarity of movement. We propose that a region adjacent to the catalytic domain serves as a mechanical transducer that determines directionality.
- Full Text
- View/download PDF
10. Albumin-bound nanoparticle (nab) paclitaxel exhibits enhanced paclitaxel tissue distribution and tumor penetration
- Author
-
Simon Zhou, Nianhang Chen, Xiping Liu, Joyoti Dey, William S. Kerwin, Maria Palmisano, Shihe Hou, Richard A. Klinghoffer, Michael Carleton, Rajesh Chopra, Yan Li, Daniel W. Pierce, and Carrie Baker Brachmann
- Subjects
Cancer Research ,Microinjections ,Paclitaxel ,Serum albumin ,Mice, Nude ,Nanoparticle ,Serum Albumin, Human ,Endosomes ,Taxane ,Pharmacology ,Toxicology ,Capillary Permeability ,nab-paclitaxel ,chemistry.chemical_compound ,Drug Delivery Systems ,Cell Line, Tumor ,Cremophor EL ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Tissue Distribution ,Pharmacology (medical) ,Tissue distribution ,Infusions, Intravenous ,Cells, Cultured ,Serum Albumin ,Nab-paclitaxel ,biology ,Albumin ,Carcinoma ,Biological Transport ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,Tubulin Modulators ,Pancreatic Neoplasms ,chemistry ,Oncology ,Cell culture ,Cancer research ,biology.protein ,Nanoparticles ,Original Article ,Endothelium, Vascular - Abstract
Purpose nab-paclitaxel demonstrates improved clinical efficacy compared with conventional Cremophor EL (CrEL)-paclitaxel in multiple tumor types. This study explored the distinctions in drug distribution between nab-paclitaxel and CrEL-paclitaxel and the underlying mechanisms. Methods Uptake and transcytosis of paclitaxel were analyzed by vascular permeability assay across human endothelial cell monolayers. The tissue penetration of paclitaxel within tumors was evaluated by local injections into tumor xenografts and quantitative image analysis. The distribution profile of paclitaxel in solid-tumor patients was assessed using pharmacokinetic modeling and simulation. Results Live imaging demonstrated that albumin and paclitaxel were present in punctae in endothelial cells and could be observed in very close proximity, suggesting cotransport. Uptake and transport of albumin, nab-paclitaxel and paclitaxel were inhibited by clinically relevant CrEL concentrations. Further, nab-paclitaxel causes greater mitotic arrest in wider area within xenografted tumors than CrEL- or dimethyl sulfoxide-paclitaxel following local microinjection, demonstrating enhanced paclitaxel penetration and uptake by albumin within tumors. Modeling of paclitaxel distribution in patients with solid tumors indicated that nab-paclitaxel is more dependent upon transporter-mediated pathways for drug distribution into tissues than CrEL-paclitaxel. The percent dose delivered to tissue via transporter-mediated pathways is predicted to be constant with nab-paclitaxel but decrease with increasing CrEL-paclitaxel dose. Conclusions Compared with CrEL-paclitaxel, nab-paclitaxel demonstrated more efficient transport across endothelial cells, greater penetration and cytotoxic induction in xenograft tumors, and enhanced extravascular distribution in patients that are attributed to carrier-mediated transport. These observations are consistent with the distinct clinical efficacy and toxicity profile of nab-paclitaxel. Electronic supplementary material The online version of this article (doi:10.1007/s00280-015-2833-5) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.