109 results on '"Judith A. Fox"'
Search Results
2. Vecabrutinib inhibits B-cell receptor signal transduction in chronic lymphocytic leukemia cell types with wild-type or mutant Bruton tyrosine kinase
- Author
-
Burcu Aslan, Stefan Edward Hubner, Judith A. Fox, Pietro Taverna, William G. Wierda, Steven M. Kornblau, and Varsha Gandhi
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
3. Phase 3 results for vosaroxin/cytarabine in the subset of patients ≥60 years old with refractory/early relapsed acute myeloid leukemia
- Author
-
Farhad Ravandi, Ellen K. Ritchie, Hamid Sayar, Jeffrey E. Lancet, Michael D. Craig, Norbert Vey, Stephen A. Strickland, Gary J. Schiller, Elias Jabbour, Arnaud Pigneux, Heinz-August Horst, Christian Récher, Virginia M. Klimek, Jorge E. Cortes, Angelo-Michele Carella, Miklos Egyed, Utz Krug, Judith A. Fox, Adam R. Craig, Renee Ward, Jennifer A. Smith, Gary Acton, Hagop M. Kantarjian, and Robert K. Stuart
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
4. A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia
- Author
-
Jeffrey E. Lancet, Gail J. Roboz, Larry D. Cripe, Glenn C. Michelson, Judith A. Fox, Richard D. Leavitt, Tianling Chen, Rachael Hawtin, Adam R. Craig, Farhad Ravandi, Michael B. Maris, Robert K. Stuart, and Judith E. Karp
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Vosaroxin is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II. This study assessed the safety and tolerability of vosaroxin plus cytarabine in patients with relapsed/refractory acute myeloid leukemia. Escalating vosaroxin doses (10-minute infusion; 10–90 mg/m2; days 1, 4) were given in combination with cytarabine on one of two schedules: schedule A (24-hour continuous intravenous infusion, 400 mg/m2/day, days 1–5) or schedule B (2-hour intravenous infusion, 1 g/m2/day, days 1–5). Following dose escalation, enrollment was expanded at the maximum tolerated dose. Of 110 patients enrolled, 108 received treatment. The maximum tolerated dose of vosaroxin was 80 mg/m2 for schedule A (dose-limiting toxicities: grade 3 bowel obstruction and stomatitis) and was not reached for schedule B (recommended phase 2 dose: 90 mg/m2). In the efficacy population (all patients in first relapse or with primary refractory disease treated with vosaroxin 80–90 mg/m2; n=69), the complete remission rate was 25% and the complete remission/complete remission with incomplete blood count recovery rate was 28%. The 30-day all-cause mortality rate was 2.5% among all patients treated at a dose of 80–90 mg/m2. Based upon these results, a phase 3 trial of vosaroxin plus cytarabine was initiated in patients with relapsed/refractory acute myeloid leukemia.
- Published
- 2015
- Full Text
- View/download PDF
5. Data from Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia
- Author
-
Roland B. Walter, Jeanmarie Guenot, Lori A. Kunkel, Judith A. Fox, Eugene A. Zhukovsky, Stefan H.J. Knackmuss, Michael Weichel, Kristina Ellwanger, Ivica Fucek, Chelsea J. Gudgeon, Kimberly H. Harrington, and Uwe Reusch
- Abstract
Purpose: Randomized studies with gemtuzumab ozogamicin have validated CD33 as a target for antigen-specific immunotherapy of acute myelogenous leukemia (AML). Here, we investigated the potential of CD33/CD3-directed tandem diabodies (TandAbs) as novel treatment approach for AML. These tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a bivalent antibody and have a molecular weight exceeding the renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs.Experimental Design: We constructed a series of TandAbs composed of anti-CD33 and anti-CD3 variable domains of diverse binding affinities and profiled their functional properties in CD33+ human leukemia cell lines, xenograft models, and AML patient samples.Results: Our studies demonstrated that several CD33/CD3 TandAbs could induce potent, dose-dependent cytolysis of CD33+ AML cell lines. This effect was modulated by the effector-to-target cell ratio and strictly required the presence of T cells. Activation and proliferation of T cells and maximal AML cell cytolysis correlated with high avidity to both CD33 and CD3. High-avidity TandAbs were broadly active in primary specimens from patients with newly diagnosed or relapsed/refractory AML in vitro, with cytotoxic properties independent of CD33 receptor density and cytogenetic risk. Tumor growth delay and inhibition were observed in both prophylactic and established HL-60 xenograft models in immunodeficient mice.Conclusions: Our data show high efficacy of CD33/CD3 TandAbs in various preclinical models of human AML. Together, these findings support further study of CD33/CD3 TandAbs as novel immunotherapeutics for patients with AML. Clin Cancer Res; 22(23); 5829–38. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia
- Author
-
Roland B. Walter, Jeanmarie Guenot, Lori A. Kunkel, Judith A. Fox, Eugene A. Zhukovsky, Stefan H.J. Knackmuss, Michael Weichel, Kristina Ellwanger, Ivica Fucek, Chelsea J. Gudgeon, Kimberly H. Harrington, and Uwe Reusch
- Abstract
Flow cytometry analysis of for AML cell cytotoxicity determinations. Flow cytometry analysis of cells from one healthy donor T-cell aliquot and one representative AML cell line (HL- 60) and primary AML specimen (AMP002) illustrating the strategy pursued to determine antibody-induced cytotoxicity.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia
- Author
-
Roland B. Walter, Jeanmarie Guenot, Lori A. Kunkel, Judith A. Fox, Eugene A. Zhukovsky, Stefan H.J. Knackmuss, Michael Weichel, Kristina Ellwanger, Ivica Fucek, Chelsea J. Gudgeon, Kimberly H. Harrington, and Uwe Reusch
- Abstract
Proliferation assays with PBMCs and T-cell cultures. Incorporation of BrdU in proliferating cells over a four-day incubation quantified using a BrdU ELISA as a function of increasing TandAb concentration.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 3 from Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia
- Author
-
Roland B. Walter, Jeanmarie Guenot, Lori A. Kunkel, Judith A. Fox, Eugene A. Zhukovsky, Stefan H.J. Knackmuss, Michael Weichel, Kristina Ellwanger, Ivica Fucek, Chelsea J. Gudgeon, Kimberly H. Harrington, and Uwe Reusch
- Abstract
Correlation of CD3 and CD33 affinity with EC50 in T-cell proliferation assays. Data points from proliferation assays shown in Supplemental Figure 2 were plotted as a function of KD on T-cells or CD33+ cells.
- Published
- 2023
- Full Text
- View/download PDF
9. Impact of Being a Peer Recovery Specialist on Work and Personal Life: Implications for Training and Supervision
- Author
-
Amanda L. Roy, Annemarie Vaccaro, Sara D. Cottrill, Marie C. Tate, Emma Lund, Meinca Pinchinat, L A R Stein, and Judith B Fox
- Subjects
Service (business) ,Medical education ,Health (social science) ,Service delivery framework ,Public Health, Environmental and Occupational Health ,Personal life ,Focus group ,Mental health ,030227 psychiatry ,03 medical and health sciences ,Psychiatry and Mental health ,0302 clinical medicine ,Consistency (negotiation) ,Resource (project management) ,Work (electrical) ,030212 general & internal medicine ,Psychology - Abstract
Peer recovery specialists are an important resource in community mental health settings. This study, which was part of a larger statewide assessment, evaluates how the role impacts work and personal lives of peers, with implications for improving the training and supervision of this service. The importance of peer work has been investigated through client outcomes, however less work has investigated outcomes on peers themselves, which impacts the work force and service delivery. Nine focus groups were conducted with peer recovery specialists. A two-stage qualitative analysis led to two overarching themes, work and personal, and six subthemes. Findings suggest being a peer presents unique benefits and challenges in work and personal life. Peers benefit from more training and supervision, consistency within the role, and maintaining boundaries. Additionally, work environment roles may be improved by attention to needs of supervisors in terms of skills for effective supervision and clarification of supervisory roles.
- Published
- 2021
- Full Text
- View/download PDF
10. Immunodepletion of MDSC by AMV564, a novel bivalent, bispecific CD33/CD3 T cell engager, ex vivo in MDS and melanoma
- Author
-
Pingyan Cheng, Xianghong Chen, Robert Dalton, Alexandra Calescibetta, Tina So, Danielle Gilvary, Grace Ward, Victoria Smith, Sterling Eckard, Judith A. Fox, Jeanmarie Guenot, Joseph Markowitz, John L. Cleveland, Kenneth L. Wright, Alan F. List, Sheng Wei, and Erika A. Eksioglu
- Subjects
Pharmacology ,Myeloid-Derived Suppressor Cells ,T-Lymphocytes ,Sialic Acid Binding Ig-like Lectin 3 ,Antineoplastic Agents ,Myelodysplastic Syndromes ,Drug Discovery ,Antibodies, Bispecific ,Genetics ,Molecular Medicine ,Animals ,Humans ,Molecular Biology ,Melanoma - Abstract
We have reported previously that CD33
- Published
- 2021
11. Evaluation of vecabrutinib as a model for noncovalent BTK/ITK inhibition for treatment of chronic lymphocytic leukemia
- Author
-
Eugen Tausch, Stephan Stilgenbauer, Annika Müller, Felix Seyfried, Sascha Endres, Martina Seiffert, Rashmi Priyadharshini Dheenadayalan, Martin Wist, Klaus-Michael Debatin, Daniel Mertens, Billy Michael Chelliah Jebaraj, Judith A. Fox, Lueder H. Meyer, Philipp M. Roessner, Claudia Walliser, Peter Gierschik, Jialei Qi, and Pietro Taverna
- Subjects
Models, Molecular ,Adoptive cell transfer ,Chronic lymphocytic leukemia ,Immunology ,Biochemistry ,chemistry.chemical_compound ,immune system diseases ,In vivo ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Agammaglobulinaemia Tyrosine Kinase ,Bruton's tyrosine kinase ,Animals ,Humans ,Protein Kinase Inhibitors ,Tumor microenvironment ,biology ,Kinase ,Chemistry ,Venetoclax ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Tumor Burden ,Mice, Inbred C57BL ,Ibrutinib ,biology.protein ,Cancer research ,Female - Abstract
Covalent Bruton tyrosine kinase (BTK) inhibitors, such as ibrutinib, have proven to be highly beneficial in the treatment of chronic lymphocytic leukemia (CLL). Interestingly, the off-target inhibition of IL-2-inducible T-cell kinase (ITK) by ibrutinib may also play a role in modulating the tumor microenvironment, potentially enhancing the treatment benefit. However, resistance to covalently binding BTK inhibitors can develop as the result of a mutation in cysteine 481 of BTK (C481S), which prevents irreversible binding of the drugs. In the present study we performed preclinical characterization of vecabrutinib, a next-generation noncovalent BTK inhibitor that has ITK-inhibitory properties similar to those of ibrutinib. Unlike ibrutinib and other covalent BTK inhibitors, vecabrutinib showed retention of the inhibitory effect on C481S BTK mutants in vitro, similar to that of wild-type BTK. In the murine Eμ-TCL1 adoptive transfer model, vecabrutinib reduced tumor burden and significantly improved survival. Vecabrutinib treatment led to a decrease in CD8+ effector and memory T-cell populations, whereas the naive populations were increased. Of importance, vecabrutinib treatment significantly reduced the frequency of regulatory CD4+ T cells in vivo. Unlike ibrutinib, vecabrutinib treatment showed minimal adverse impact on the activation and proliferation of isolated T cells. Lastly, combination treatment with vecabrutinib and venetoclax augmented treatment efficacy, significantly improved survival, and led to favorable reprogramming of the microenvironment in the murine Eμ-TCL1 model. Thus, noncovalent BTK/ITK inhibitors, such as vecabrutinib, may be efficacious in C481S BTK mutant CLL while preserving the T-cell immunomodulatory function of ibrutinib.
- Published
- 2021
12. Impact of Being a Peer Recovery Specialist on Work and Personal Life: Implications for Training and Supervision
- Author
-
Marie C, Tate, Amanda, Roy, Meinca, Pinchinat, Emma, Lund, Judith B, Fox, Sara, Cottrill, Annemarie, Vaccaro, and L A R, Stein
- Subjects
Workforce ,Humans ,Focus Groups ,Peer Group ,Specialization - Abstract
Peer recovery specialists are an important resource in community mental health settings. This study, which was part of a larger statewide assessment, evaluates how the role impacts work and personal lives of peers, with implications for improving the training and supervision of this service. The importance of peer work has been investigated through client outcomes, however less work has investigated outcomes on peers themselves, which impacts the work force and service delivery. Nine focus groups were conducted with peer recovery specialists. A two-stage qualitative analysis led to two overarching themes, work and personal, and six subthemes. Findings suggest being a peer presents unique benefits and challenges in work and personal life. Peers benefit from more training and supervision, consistency within the role, and maintaining boundaries. Additionally, work environment roles may be improved by attention to needs of supervisors in terms of skills for effective supervision and clarification of supervisory roles.
- Published
- 2020
13. Favorable Modulation of Chimeric Antigen Receptor T Cells Safety and Efficacy By the Non-Covalent BTK Inhibitor Vecabrutinib
- Author
-
Elizabeth L. Siegler, Lionel A. Kankeu Fonkoua, Reona Sakemura, Neil E. Kay, Evandro D. Bezerra, Carli M. Stewart, Gloria Olivier, Caludia Manriquez-Roman, Sameer A. Parikh, Kendall J. Schick, Mohamad M. Adada, Michelle J. Cox, Judith A. Fox, Wei Ding, Mehrdad Hefazi, Michael W. Ruff, Ismail Can, Susan L. Slager, Saad S. Kenderian, Erin E. Tapper, Pietro Taverna, and Ekene J. Ogbodo
- Subjects
Transplantation ,biology ,Chemistry ,Non covalent ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Chimeric antigen receptor ,Cell biology ,biology.protein ,Bruton's tyrosine kinase ,Molecular Medicine ,Immunology and Allergy ,health care economics and organizations - Abstract
CD19-targeted chimeric antigen receptor T cell (CART19) therapy has been remarkably successful in treating a subset of patients with hematological malignancies. However, it is associated with significant toxicities, including cytokine release syndrome (CRS) and neurotoxicity (NT). Furthermore, the rate of durable responses after CART19 therapy is low, and most patients develop resistance to the therapy. One of the predominant mechanisms for CART cell resistance is intrinsic T cell dysfunction which leads to a suboptimal CART product. The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has been shown to favorably modulate CART phenotype and function through downregulation of inhibitory receptors and enhancement of CART cell efficacy in preclinical models and early clinical studies. However, ibrutinib is an irreversible receptor tyrosine kinase (RTK) inhibitor, and thus there are concerns that it inhibits CART cell proliferation and expansion. In contrast to ibrutinib and other BTK inhibitors, vecabrutinib has recently emerged as a potent, reversible, non-covalent BTK inhibitor that can block the activity of both wild type and C481 mutant BTK as well as interleukin-2-inducible T-cell kinase (ITK). We hypothesized that treatment with vecabrutinib improves CART19 antitumor efficacy through the reversible inhibition of BTK/ITK, independent of its antitumor effect. To test this hypothesis, we assessed whether vecabrutinib potentiates CART19 cytotoxicity against CD19 + tumor cells. Killing of the CD19 + mantle cell lymphoma cell line, JeKo-1, was significantly increased when CART19 were combined with vecabrutinib (Fig. 1A). Of note, vecabrutinib did not induce antitumor activity in this model as a single agent (Fig. 1A). In addition, vecabrutinib (10 µM) treatment of CART19 resulted in enhanced antigen-specific proliferation (Fig. 1B). Next, we aimed to study the direct impact of vecabrutinib on CART19 cytokine production. We measured the levels of multiple pro-inflammatory cytokines in the supernatant of CART19 co-cultured for 3 days with JeKo-1 cells and vecabrutinib. Vecabrutinib did not impair CART19 degranulation, as measured by flow cytometric assessment of CD107a expression (Fig. 1C). Cytokines known to play a crucial part in the development of CRS, such as IL-6, IL-10, and MIP-1β were significantly downregulated in cocultures with vecabrutinib (Fig. 1D-F). We then investigated whether the addition of vecabrutinib to CART19 caused a synergistic antitumor effect in vivo. We used our established JeKo-1 lymphoma xenograft models. Here NSG mice were engrafted with CD19 + luciferase + JeKo-1 cells. Mice were then imaged for engraftment and randomized to treatment with 1) untransduced control T cells, 2) CART19 with vehicle, or 3) CART19 with vecabrutinib 50 mg/kg BID administered via oral gavage over a period of 4 weeks. Vecabrutinib combination with CART19 treatment led to sustained antitumor activity (Fig.1G) and increased CART19 proliferation (Fig.1H). Encouragingly, mice treated with vecabrutinib better maintained their body weight following CART19 infusion, suggesting possibly dampened toxicity (Fig.1I). We also measured serum cytokine levels from patients on a phase 1 clinical trial (NCT03037645) testing vecabrutinib in B cell malignancies at baseline and four weeks after treatment. We found that vecabrutinib significantly reduces the levels of multiple pro-inflammatory cytokines linked to CART cell toxicity, including MIP-1β, IP-10, and TNF-a (Fig. 1J-L), further validating our preclinical findings. Finally, we interrogated the transcriptome of CART19 co-cultured with JeKo-1 with or without vecabrutinib. Total RNA sequencing of activated CART19 highlighted a significant enhanced expression of multiple genes involved in the PI3K/AKT and Th1 pathways compared to antigen stimulated CART19 alone (Fig.1M, N). These results could explain the enhanced proliferation and cytotoxicity induced by vecabrutinib. In summary, we demonstrate for the first time that using the reversible BTK inhibitor, vecabrutinib, is a potent and a novel strategy to favorably modulate CART19 function by increasing their efficacy and decreasing toxicity. Further studies are currently underway to compare the effect of reversible versus irreversible BTK/ITK inhibition on CART functions and to further validate the mechanisms responsible for the observed effects. Figure 1 Figure 1. Disclosures Sakemura: Humanigen: Patents & Royalties. Cox: Humanigen: Patents & Royalties. Fox: Sunesis Pharmaceuticals: Current Employment. Ding: Merck: Membership on an entity's Board of Directors or advisory committees, Research Funding; Octapharma: Membership on an entity's Board of Directors or advisory committees; DTRM: Research Funding. Parikh: Pharmacyclics, MorphoSys, Janssen, AstraZeneca, TG Therapeutics, Bristol Myers Squibb, Merck, AbbVie, and Ascentage Pharma: Research Funding; Pharmacyclics, AstraZeneca, Genentech, Gilead, GlaxoSmithKline, Verastem Oncology, and AbbVie: Membership on an entity's Board of Directors or advisory committees. Kay: MEI Pharma: Research Funding; Sunesis: Research Funding; Rigel: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; CytomX Therapeutics: Membership on an entity's Board of Directors or advisory committees; TG Therapeutics: Research Funding; Juno Therapeutics: Membership on an entity's Board of Directors or advisory committees; Bristol Meyer Squib: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Behring: Membership on an entity's Board of Directors or advisory committees; Genentech: Research Funding; Morpho-sys: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Targeted Oncology: Membership on an entity's Board of Directors or advisory committees; Tolero Pharmaceuticals: Research Funding; Dava Oncology: Membership on an entity's Board of Directors or advisory committees; Acerta Pharma: Research Funding; Agios Pharm: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Oncotracker: Membership on an entity's Board of Directors or advisory committees. Kenderian: Humanigen, Inc.: Consultancy, Honoraria, Research Funding.
- Published
- 2022
- Full Text
- View/download PDF
14. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons topoisomerase II.
- Author
-
Rachael E Hawtin, David E Stockett, Jo Ann W Byl, Robert S McDowell, Tan Nguyen, Michelle R Arkin, Andrew Conroy, Wenjin Yang, Neil Osheroff, and Judith A Fox
- Subjects
Medicine ,Science - Abstract
Topoisomerase II is critical for DNA replication, transcription and chromosome segregation and is a well validated target of anti-neoplastic drugs including the anthracyclines and epipodophyllotoxins. However, these drugs are limited by common tumor resistance mechanisms and side-effect profiles. Novel topoisomerase II-targeting agents may benefit patients who prove resistant to currently available topoisomerase II-targeting drugs or encounter unacceptable toxicities. Voreloxin is an anticancer quinolone derivative, a chemical scaffold not used previously for cancer treatment. Voreloxin is completing Phase 2 clinical trials in acute myeloid leukemia and platinum-resistant ovarian cancer. This study defined voreloxin's anticancer mechanism of action as a critical component of rational clinical development informed by translational research.Biochemical and cell-based studies established that voreloxin intercalates DNA and poisons topoisomerase II, causing DNA double-strand breaks, G2 arrest, and apoptosis. Voreloxin is differentiated both structurally and mechanistically from other topoisomerase II poisons currently in use as chemotherapeutics. In cell-based studies, voreloxin poisoned topoisomerase II and caused dose-dependent, site-selective DNA fragmentation analogous to that of quinolone antibacterials in prokaryotes; in contrast etoposide, the nonintercalating epipodophyllotoxin topoisomerase II poison, caused extensive DNA fragmentation. Etoposide's activity was highly dependent on topoisomerase II while voreloxin and the intercalating anthracycline topoisomerase II poison, doxorubicin, had comparable dependence on this enzyme for inducing G2 arrest. Mechanistic interrogation with voreloxin analogs revealed that intercalation is required for voreloxin's activity; a nonintercalating analog did not inhibit proliferation or induce G2 arrest, while an analog with enhanced intercalation was 9.5-fold more potent.As a first-in-class anticancer quinolone derivative, voreloxin is a toposiomerase II-targeting agent with a unique mechanistic signature. A detailed understanding of voreloxin's molecular mechanism, in combination with its evolving clinical profile, may advance our understanding of structure-activity relationships to develop safer and more effective topoisomerase II-targeted therapies for the treatment of cancer.
- Published
- 2010
- Full Text
- View/download PDF
15. Efficacy of Vecabrutinib Treatment in a Murine Model of Sclerodermatous Graft-Versus-Host-Disease
- Author
-
Pietro Taverna, Melanie Mediavilla-Varela, Wael Gamal, Eva Sahakian, Judith A. Fox, Angimar Uriepero, Javier Pinilla Ibarz, and Kamira Maharaj
- Subjects
Graft-versus-host disease ,business.industry ,Murine model ,Immunology ,medicine ,Cell Biology ,Hematology ,medicine.disease ,business ,Biochemistry - Abstract
Introduction. Chronic graft-versus-host disease (cGVHD) can manifest as a complication in patients following allogeneic hematopoietic stem cell transplant resulting in morbidity and mortality. Effective treatment strategies for cGVHD are currently lacking. Ibrutinib, an irreversible BTK inhibitor with activity against several other Tec-family kinases, has been clinically developed for cGVHD treatment due to regulation of pathogenetic B cells and T-cell subsets. Vecabrutinib is a more selective, reversible inhibitor of BTK with a distinct kinase domain interaction, which could result in differentiated safety and activity profiles compared to ibrutinib. Vecabrutinib has the capacity to overcome the C481S mutation that mediates resistance to ibrutinib. Vecabrutinib also also demonstrates activity against ITK, which is expressed in T cells. In this study, we investigated the activity and immune modulation of vecabrutinib treatment in a murine model of sclerodermatous cGVHD. Ibrutinib was utilized for comparison. Methods. A murine model of sclerodermatous cGVHD was initiated by adoptive transfer of T-cell depleted bone marrow plus whole splenocytes from B10.D2 donors into BALB/c recipients that were previously subjected to sub-lethal irradiation. Total bone marrow and irradiation only groups were included as controls. Mice with established cGVHD characterized by weight loss and skin irritation symptoms were treated with vecabrutinib once daily at 50mg/kg by oral gavage or ibrutinib at 30mg/kg in drinking water 5 days per week for approximately 3 weeks beginning on day 27 post-adoptive transfer and ending on day 45 (n=10 mice per group). Body weight and clinical symptoms (appearance, activity, skin symptoms, diarrhea, conjunctivitis) were measured throughout the study. Immunophenotyping for B cells and T cells was performed on spleens collected from euthanized mice by flow cytometry at two timepoints (day 40 during treatment and on day 76 post-treatment). Levels of circulating immunoglobulins were measured by multiplex cytokine assay at both timepoints. Results. Clinical symptoms, including appearance, activity, skin irritation, redness, alopecia and diarrhea were significantly reduced in vecabrutinib- and ibrutinib-treated groups. Furthermore, there was a trend toward a more favorable clinical score overall for the vecabrutinib-treated group, however no statistical difference was observed compared to ibrutinib possibly due to small sample size. Weight loss was slightly elevated during vecabrutinib and ibrutinib treatment compared to vehicle (trend), however mice recovered body weight post-treatment and continued to maintain benefit. On day 40 (during treatment) and day 76 (post-treatment) groups of mice were euthanized for immunophenotyping analysis utilizing a 22-color flow cytometry panel. During treatment, both vecabrutinib and ibrutinib-treated mice retained total B cell numbers but exhibited reduced B-cell activation, proliferation, and number of B220+ CD138+ plasma cells. In addition, B cells secreted less IL-10, and IL-4/5. Expression of antigen-presentation molecules CD80 and CD86 on B cells were unchanged. Total CD3+ T cells, activated and proliferating CD4+ and CD8+ T cells, and cytotoxic granzyme-B+ CD8+ T cells were reduced in treated mice. Interestingly, CD4+ CD25+ FoxP3+ Treg number, expression of PD-1 on Tregs and CD4+ CXCR5+ PD-1+ T follicular helper cells were also reduced in both treatment groups. In addition, there were globally reduced numbers of cytokine-secreting CD4+ cells but no differences in Th1/Th2 or Th17/Treg ratios were observed. Post-treatment, proliferation and cytokine secretion of B cells and T cells were still lowered but less impaired than during treatment. Tregs and PD-1 expression were still reduced post-treatment. Finally, circulating levels of IgA were reduced during and post treatment in vecabrutinib-treated mice compared to vehicle, while IgG1, IgG2b were reduced in both treated groups. No changes in IgM levels were observed in either treatment group. In conclusion, vecabrutinib treatment demonstrated efficacy and beneficially regulated B cell and T cell immune subsets in a preclinical murine model of sclerodermatous cGVHD. Studies to further evaluate differences between vecabrutinib and ibrutinib treatment are ongoing. Figure 1 Figure 1. Disclosures Fox: Sunesis Pharmaceuticals: Current Employment. Taverna: Sunesis Pharmaceuticals: Current Employment. Pinilla Ibarz: Sellas: Other: ), patents/royalties/other intellectual property; MEI, Sunesis: Research Funding; AbbVie, Janssen, AstraZeneca, Takeda: Speakers Bureau; AbbVie, Janssen, AstraZeneca, Novartis, TG Therapeutics, Takeda: Consultancy, Other: Advisory.
- Published
- 2021
- Full Text
- View/download PDF
16. Zombie Mortgages and Abandoned Properties
- Author
-
Linda E. Fisher and Judith L. Fox
- Subjects
Zombie ,Economic history ,Economics - Published
- 2019
- Full Text
- View/download PDF
17. The Breakdown of Mortgage Servicing and Loss Mitigation
- Author
-
Linda E. Fisher and Judith L. Fox
- Subjects
Natural resource economics ,Business ,Loss mitigation - Published
- 2019
- Full Text
- View/download PDF
18. PDK1 inhibitor SNS-510 shows synergy with targeted cancer therapies in solid tumor and hematologic cancer models
- Author
-
P. Taverna, S. Hansen, and Judith A. Fox
- Subjects
Cancer Research ,Hematologic cancer ,Oncology ,business.industry ,Cancer research ,Medicine ,Cancer ,business ,medicine.disease ,Solid tumor - Published
- 2020
- Full Text
- View/download PDF
19. The Foreclosure Echo: How the Hardest Hit Have Been Left Out of the Economic Recovery (Introduction)
- Author
-
Judith L. Fox and Linda E. Fisher
- Subjects
Legal research ,Inequality ,media_common.quotation_subject ,Political science ,Political economy ,Economic recovery ,Echo (computing) ,Financial crisis ,Public institution ,Table of contents ,Foreclosure ,media_common - Abstract
This paper includes the Table of Contents and Introduction to a book recently published by Cambridge University Press: It tells the story of the foreclosure crisis from a new perspective – that of ordinary people who experienced it. Using actual experiences – often examined through a legal lens – supplemented by economic, social science and legal research, The Foreclosure Echo explains how people experienced the crisis and how their lenders and public institutions let them down. The book also details the lingering effects of the crisis – such as vacant and abandoned buildings – and how these effects have magnified inequality. Finally, the book suggests reforms that could help avoid another crisis.
- Published
- 2019
- Full Text
- View/download PDF
20. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study
- Author
-
Ellen K. Ritchie, Judith A. Fox, Cyrus R. Mehta, Olatoyosi Odenike, Andrew H. Wei, Renee Ward, Gail J. Roboz, Michael Heuser, Norbert Vey, Gary Acton, Stephen A. Strickland, Elias Jabbour, Jeffrey E. Lancet, Arnaud Pigneux, Adam R. Craig, Donna E. Hogge, Gary J. Schiller, Gianluca Gaidano, Michael Craig, Violaine Havelange, Bayard L. Powell, Lloyd E. Damon, Xavier Thomas, Heinz A. Horst, Jennifer A. Smith, Christian Recher, Harry P. Erba, Robert K. Stuart, Angelo Michele Carella, Farhad Ravandi, Hagop M. Kantarjian, Jorge E. Cortes, Johan Maertens, Hans Günter Derigs, Virginia M. Klimek, Hamid Sayar, UCL - SSS/DDUV - Institut de Duve, and UCL - (SLuc) Service d'hématologie
- Subjects
Myeloid ,Male ,Phases of clinical research ,Kaplan-Meier Estimate ,Vosaroxin ,Gastroenterology ,chemistry.chemical_compound ,Antineoplastic Combined Chemotherapy Protocols ,80 and over ,Clinical endpoint ,Cancer ,Aged, 80 and over ,Leukemia ,Remission Induction ,Cytarabine ,Hematology ,Middle Aged ,Leukemia, Myeloid, Acute ,Treatment Outcome ,Local ,Oncology ,6.1 Pharmaceuticals ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Acute ,Neutropenia ,Placebo ,Article ,Disease-Free Survival ,Double-Blind Method ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Naphthyridines ,Aged ,Intention-to-treat analysis ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Surgery ,Thiazoles ,Neoplasm Recurrence ,chemistry ,Neoplasm Recurrence, Local ,business ,Febrile neutropenia - Abstract
BACKGROUND: Safe and effective treatments are urgently needed for patients with relapsed or refractory acute myeloid leukaemia. We investigated the efficacy and safety of vosaroxin, a first-in-class anticancer quinolone derivative, plus cytarabine in patients with relapsed or refractory acute myeloid leukaemia. METHODS: This phase 3, double-blind, placebo-controlled trial was undertaken at 101 international sites. Eligible patients with acute myeloid leukaemia were aged 18 years of age or older and had refractory disease or were in first relapse after one or two cycles of previous induction chemotherapy, including at least one cycle of anthracycline (or anthracenedione) plus cytarabine. Patients were randomly assigned 1:1 to vosaroxin (90 mg/m(2) intravenously on days 1 and 4 in a first cycle; 70 mg/m(2) in subsequent cycles) plus cytarabine (1 g/m(2) intravenously on days 1-5) or placebo plus cytarabine through a central interactive voice system with a permuted block procedure stratified by disease status, age, and geographical location. All participants were masked to treatment assignment. The primary efficacy endpoint was overall survival and the primary safety endpoint was 30-day and 60-day all-cause mortality. Efficacy analyses were done by intention to treat; safety analyses included all treated patients. This study is registered with ClinicalTrials.gov, number NCT01191801. FINDINGS: Between Dec 17, 2010, and Sept 25, 2013, 711 patients were randomly assigned to vosaroxin plus cytarabine (n=356) or placebo plus cytarabine (n=355). At the final analysis, median overall survival was 7·5 months (95% CI 6·4-8·5) in the vosaroxin plus cytarabine group and 6·1 months (5·2-7·1) in the placebo plus cytarabine group (hazard ratio 0·87, 95% CI 0·73-1·02; unstratified log-rank p=0·061; stratified p=0·024). A higher proportion of patients achieved complete remission in the vosaroxin plus cytarabine group than in the placebo plus cytarabine group (107 [30%] of 356 patients vs 58 [16%] of 355 patients, p
- Published
- 2015
- Full Text
- View/download PDF
21. Iron(<scp>iii</scp>)-binding of the anticancer agents doxorubicin and vosaroxin
- Author
-
Chris Orvig, Gene Jamieson, Jacqueline F. Cawthray, Judith A. Fox, and Katja Dralle Mjos
- Subjects
Models, Molecular ,Tris ,medicine.drug_class ,Stereochemistry ,Iron ,Molecular Conformation ,Antineoplastic Agents ,Gallium ,Vosaroxin ,Redox ,Medicinal chemistry ,Inorganic Chemistry ,chemistry.chemical_compound ,Drug Stability ,Organometallic Compounds ,medicine ,Doxorubicin ,Naphthyridines ,chemistry.chemical_classification ,biology ,Topoisomerase ,Quinolone ,Thiazoles ,Enzyme ,Mechanism of action ,chemistry ,biology.protein ,medicine.symptom ,medicine.drug - Abstract
The Fe(iii)-binding constant of vosaroxin, an anticancer quinolone derivative, has been determined spectrophotometrically and compared with the analogous Fe(iii) complex formed with doxorubicin. The in vivo metabolic stability and iron coordination properties of the quinolones compared to the anthracylines may provide significant benefit to cardiovascular safety. The mechanism of action of both molecules target the topoisomerase II enzyme. Both doxorubicin (Hdox, log βFeL3 = 33.41, pM = 17.0) and vosaroxin (Hvox, log βFeL3 = 33.80(3), pM = 15.9) bind iron(iii) with comparable strength; at physiological pH however, [Fe(vox)3] is the predominant species in contrast to a mixture of species observed for the Fe:dox system. Iron(iii) nitrate and gallium(iii) nitrate at a 1 : 3 ratio with vosaroxin formed stable tris(vosaroxacino)-iron(iii) and tris(vosaroxino)gallium(iii) complexes that were isolated and characterized. Their redox behavior was studied by CV, and their stereochemistry was further explored in temperature dependent (1)H NMR studies. The molecular pharmacology of their interaction with iron(iii) may be one possible differentiation in the safety profile of quinolones compared to anthracyclines in relation to cardiotoxicity.
- Published
- 2015
- Full Text
- View/download PDF
22. <scp>REVEAL</scp> ‐1, a phase 2 dose regimen optimization study of vosaroxin in older poor‐risk patients with previously untreated acute myeloid leukaemia
- Author
-
Maureen A. Cooper, Gautam Borthakur, Larry D. Cripe, James R. Mason, Michael B. Maris, Richard Stone, Stephen A. Strickland, Shaker R. Dakhil, Farhad Ravandi, Paul J. Shami, Robert K. Stuart, Richard D. Leavitt, Glenn Michelson, Francesco Turturro, Judith A. Fox, Luciano J. Costa, and Wendy Stock
- Subjects
Male ,medicine.medical_specialty ,Population ,Phases of clinical research ,Antineoplastic Agents ,Neutropenia ,elderly ,Vosaroxin ,Gastroenterology ,Drug Administration Schedule ,chemistry.chemical_compound ,Internal medicine ,medicine ,Humans ,acute myeloid leukaemia ,Naphthyridines ,Infusions, Intravenous ,Adverse effect ,education ,Survival analysis ,Aged ,Aged, 80 and over ,education.field_of_study ,Dose-Response Relationship, Drug ,Haematological Malignancy ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Surgery ,Leukemia, Myeloid, Acute ,Thiazoles ,Regimen ,Treatment Outcome ,chemistry ,Female ,newly diagnosed ,business ,vosaroxin ,topoisomerase-II inhibitor ,Febrile neutropenia ,Research Paper - Abstract
This phase 2 study (N = 116) evaluated single-agent vosaroxin, a first-in-class anticancer quinolone derivative, in patients ≥60 years of age with previously untreated unfavourable prognosis acute myeloid leukaemia. Dose regimen optimization was explored in sequential cohorts (A: 72 mg/m2 d 1, 8, 15; B: 72 mg/m2 d 1, 8; C: 72 mg/m2 or 90 mg/m2 d 1, 4). The primary endpoint was combined complete remission rate (complete remission [CR] plus CR with incomplete platelet recovery [CRp]). Common (>20%) grade ≥3 adverse events were thrombocytopenia, febrile neutropenia, anaemia, neutropenia, sepsis, pneumonia, stomatitis and hypokalaemia. Overall CR and CR/CRp rates were 29% and 32%; median overall survival (OS) was 7·0 months; 1-year OS was 34%. Schedule C (72 mg/m2) had the most favourable safety and efficacy profile, with faster haematological recovery (median 27 d) and lowest incidence of aggregate sepsis (24%) and 30-d (7%) and 60-d (17%) all-cause mortality; at this dose and schedule, CR and CR/CRp rates were 31% and 35%, median OS was 7·7 months and 1-year OS was 38%. Overall, vosaroxin resulted in low early mortality and an encouraging response rate; vosaroxin 72 mg/m2 d 1, 4 is recommended for further study in this population. Registered at www.clinicaltrials.gov: #{"type":"clinical-trial","attrs":{"text":"NCT00607997","term_id":"NCT00607997"}}NCT00607997.
- Published
- 2014
- Full Text
- View/download PDF
23. PS1148 PRELIMINARY RESULTS OF A PHASE 1B/2 DOSE-ESCALATION AND COHORT-EXPANSION STUDY OF THE NONCOVALENT, REVERSIBLE BRUTON'S TYROSINE KINASE INHIBITOR (BTKI), VECABRUTINIB, IN B-CELL MALIGNANCIES
- Author
-
P. Taverna, M.S. Davids, John M. Pagel, Anthony R. Mato, J.R. Brown, M.Y. Choi, Gary Acton, W.G. Wierda, Renee Ward, Judith A. Fox, Susan O'Brien, R.R. Furman, J.N. Allan, J. Pinilla-Ibarz, and K. Patel
- Subjects
medicine.anatomical_structure ,business.industry ,Cohort ,Cancer research ,Dose escalation ,Medicine ,Hematology ,business ,B cell ,Bruton's tyrosine kinase inhibitor - Published
- 2019
- Full Text
- View/download PDF
24. Enhancement of radiosensitivity by the novel anticancer quinolone derivative vosaroxin in preclinical glioblastoma models
- Author
-
Flora Vitale, Luca Ventura, Francesco Marampon, Claudia Mattei, Claudio Festuccia, Antonella Vetuschi, Giovanni Luca Gravina, Judith A. Fox, Ernesto Di Cesare, Giulia Rossi, Alessandro Colapietro, and Andrea Mancini
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Apoptosis ,double-strand breaks ,glioblastoma ,radiotherapy ,topoisomerase II ,vosaroxin ,Radiation Tolerance ,Vosaroxin ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Leukocytes ,biology ,Topoisomerase II ,Tumor Burden ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Research Paper ,medicine.drug ,Programmed cell death ,Cell Survival ,Double-strand breaks ,Glioblastoma ,Radiotherapy ,Antineoplastic Agents ,Necrosis ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiosensitivity ,Naphthyridines ,Temozolomide ,Dose-Response Relationship, Drug ,business.industry ,Topoisomerase ,Dose-Response Relationship, Radiation ,Xenograft Model Antitumor Assays ,Radiation therapy ,Disease Models, Animal ,Thiazoles ,030104 developmental biology ,chemistry ,Immunology ,biology.protein ,Cancer research ,business ,Biomarkers - Abstract
// Giovanni Luca Gravina 1, 2 , Andrea Mancini 2 , Claudia Mattei 3 , Flora Vitale 3 , Francesco Marampon 2 , Alessandro Colapietro 2 , Giulia Rossi 2 , Luca Ventura 3 , Antonella Vetuschi 4 , Ernesto Di Cesare 1 , Judith A. Fox 5 , Claudio Festuccia 2 1 Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L’Aquila, L’Aquila, Italy 2 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, L’Aquila, Italy 3 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Neurosciences, University of L’Aquila, L’Aquila, Italy 4 Department of Biotechnological and Applied Clinical Sciences, Chair of Human Anatomy, University of L’Aquila, L’Aquila, Italy 5 Sunesis Pharmaceuticals Inc., South San Francisco, CA, USA Correspondence to: Claudio Festuccia, email: claudio.festuccia@univaq.it Keywords: glioblastoma, topoisomerase II, vosaroxin, double-strand breaks, radiotherapy Received: December 14, 2016 Accepted: March 03, 2017 Published: March 13, 2017 ABSTRACT Purpose: Glioblastoma multiforme (GBM) is the most aggressive brain tumor. The activity of vosaroxin, a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, was investigated in GBM preclinical models as a single agent and combined with radiotherapy (RT). Results: Vosaroxin showed antitumor activity in clonogenic survival assays, with IC 50 of 10−100 nM, and demonstrated radiosensitization. Combined treatments exhibited significantly higher γH2Ax levels compared with controls. In xenograft models, vosaroxin reduced tumor growth and showed enhanced activity with RT; vosaroxin/RT combined was more effective than temozolomide/RT. Vosaroxin/RT triggered rapid and massive cell death with characteristics of necrosis. A minor proportion of treated cells underwent caspase-dependent apoptosis, in agreement with in vitro results. Vosaroxin/RT inhibited RT-induced autophagy, increasing necrosis. This was associated with increased recruitment of granulocytes, monocytes, and undifferentiated bone marrow–derived lymphoid cells. Pharmacokinetic analyses revealed adequate blood-brain penetration of vosaroxin. Vosaroxin/RT increased disease-free survival (DFS) and overall survival (OS) significantly compared with RT, vosaroxin alone, temozolomide, and temozolomide/RT in the U251-luciferase orthotopic model. Materials and Methods: Cellular, molecular, and antiproliferative effects of vosaroxin alone or combined with RT were evaluated in 13 GBM cell lines. Tumor growth delay was determined in U87MG, U251, and T98G xenograft mouse models. (DFS) and (OS) were assessed in orthotopic intrabrain models using luciferase-transfected U251 cells by bioluminescence and magnetic resonance imaging. Conclusions: Vosaroxin demonstrated significant activity in vitro and in vivo in GBM models, and showed additive/synergistic activity when combined with RT in O6-methylguanine methyltransferase-negative and -positive cell lines.
- Published
- 2017
25. [Untitled]
- Author
-
Mark D. Rosen, Judith A. Fox, Angela Allen, Craig S. Anderson, and Liz Turner
- Subjects
Medical education ,business.industry ,Bedside ultrasound ,Medicine ,Critical Care and Intensive Care Medicine ,business ,Curriculum - Published
- 2012
- Full Text
- View/download PDF
26. AMV564, a Bivalent Bispecific (2×2) CD33/CD3 T-Cell Engager, Is Active and Improves Survival in a Mouse Model of Acute Myeloid Leukemia
- Author
-
Julie Ritchey, Jeanmarie Guenot, Linda Eissenberg, Michael P. Rettig, Judith A. Fox, and John F. DiPersio
- Subjects
0301 basic medicine ,Severe combined immunodeficiency ,biology ,business.industry ,medicine.medical_treatment ,T cell ,CD3 ,Immunology ,CD33 ,Myeloid leukemia ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,medicine ,biology.protein ,Cytotoxic T cell ,Bone marrow ,business - Abstract
Background: AMV564 is a novel bivalent, bispecific (2x2) CD33/CD3 targeted immunotherapy that binds both CD33 and the invariant CD3ε on T-cell receptors with strong avidity, thus creating an immune synapse between CD33-expressing cells and T cells, initiating T-cell directed lysis of CD33 expressing cells, and inducing expansion, differentiation and proliferation of T cells. AMV564 is being evaluated in clinical trials for patients with acute myeloid leukemia (AML, NCT03144245). Previously, we demonstrated that the parent molecule, T564, had specific, T-cell mediated cytotoxic activity against a KG-1 CD33+ cell line in vitro and eliminated blasts in an autologous AML patient-derived xenograft mouse model (Eissenberg, et al. 2015 ASCO). Here we investigated factors that may contribute to antileukemic activity of AMV564 in another, more aggressive, preclinical model of AML. Methods: Disseminated activity studies in NOD scid gamma mice (NSG) were performed by injecting MOLM13 AML cells transduced with click beetle red luciferase and green fluorescent protein (MOLM13-CG) in the tail vein. In a series of experiments, AMV564-mediated clearance of MOLM13-CG cells and overall mouse survival was determined. Tumor cells (either 3.3 x 103 or 1 x 105) were engrafted in non-conditioned NSG mice for 3-7 days. Variables included dose of AMV564 (0.5 to 25 mcg, i.v.), number of AMV564 cycles (1 or 2) and cycle length (4-5 days), number of T cell injections (1 or 2), and total number of human T cells administered (0.2 to 1.2 x 107). Tumor burden was serially measured by bioluminescence (BLI) and survival measured for 51 days. Results: Mouse survival was greatest when AMV564 was injected along with a total number of T cells ≥ 8 x 106 regardless of the number of T cell injections or their timing relative to tumor injection. AMV564 mediated up to a 3-fold increase in survival time compared to untreated mice. In the experiment shown in Figure 1, untreated mice died at day 28, yet about half of the treated mice were still alive when the experiment was terminated on day 51. In the absence of AMV564 (i.e., T cell treatment only), animal survival and tumor burden were similar to that of untreated animals. In the same experiment we administered, AMV564 daily from days 7-11 after MOLM13-CG infusion along with one injection of 8 x 106 human T cells on day 7, then measured tumor burden by BLI signal (Figure 2). By day 14 we observed a median 4.0 log or 4.8 log reduction in tumor using 5 or 25 mcg AMV564. In fact by day 14, 4 of 10 mice treated with 25 mcg AMV564 and 2 of 10 treated with 5 mcg had no detectable signal above background. No rebound in tumor burden occurred in these mice throughout the 51-day study. Conclusions: AMV564 is a potent and effective immunotherapy against an aggressive human AML cell line in NSG mice. AMV564 very effectively prolonged survival and dramatically reduced tumor in the bone marrow and peripheral blood. These results are consistent with our previous preclinical data with primary AML and support the testing of this reagent in patients with relapsed and refractory AML in the clinic. Disclosures Eissenberg: Amphivena Therapeutics: Research Funding; Novimmune: Research Funding. Rettig:Novimmune: Research Funding; Amphivena Therapeutics: Research Funding. Fox:Sunesis Pharmaceuticals: Employment; Amphivena Therapeutics: Employment. Guenot:Amphivena Therapeutics, Inc: Employment.
- Published
- 2018
- Full Text
- View/download PDF
27. Vecabrutinib Is Efficacious In Vivo in a Preclinical CLL Adoptive Transfer Model
- Author
-
Eugen Tausch, Stephan Stilgenbauer, Annika Scheffold, Billy Michael Chelliah Jebaraj, Judith A. Fox, and Pietro Taverna
- Subjects
0301 basic medicine ,Oncology ,Adoptive cell transfer ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Immunology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Medicine ,Bruton's tyrosine kinase ,IL-2 receptor ,health care economics and organizations ,biology ,business.industry ,FOXP3 ,Cell Biology ,Hematology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,biology.protein ,Bone marrow ,CD5 ,business - Abstract
B cell receptor signaling (BCR) in chronic lymphocytic leukemia (CLL) drives tumor cell proliferation and survival. Inhibition of Bruton's tyrosine kinase (BTK), a key enzyme in the BCR pathway, has proved to be efficacious even in poor-risk and chemo-refractory patients. However resistance to the BTK inhibitor ibrutinib has been shown to emerge in a subset of CLL patients. Of importance, the C481S BTK mutation conferred resistance by preventing the covalent binding of ibrutinib to its target cysteine 481 in BTK. Vecabrutinib (formerly known as SNS-062, a succinate salt) is a novel, highly potent, next generation noncovalent BTK inhibitor which demonstrated biochemical and cellular activity against C481S BTK mutant in vitro. However, the efficacy of vecabrutinib and its impact on the T-cell microenvironment has not been studied in in vivo preclinical CLL models. In the present study, the efficacy of vecabrutinib was investigated using the Eµ-TCL1 adoptive transfer model. Mice were randomized to treatment with either 40mg/kg vecabrutinib succinate, twice daily by oral gavage (n=6) or vehicle control (n=6). The mice were sacrificed after 2 weeks of treatment and changes in tumor burden as well as alterations in T-cell microenvironment were analysed in detail. Treatment with vecabrutinib decreased tumor burden as observed by a significant decrease in WBC count (36.5 vs. 17.1 giga/L; P=0.002), spleen weight (median 0.56g vs. 0.31g; P=0.005) and liver weight (median 1.5g vs. 1.2g; P=0.005) compared to vehicle treatment. Correspondingly, the CD5+ CD19+ tumor cells were significantly decreased in blood (P=0.002) and spleen (P=0.002) while no significant difference was observed in bone marrow (P=0.818) upon treatment with vecabrutinib. Since BTK inhibition is known to reshape the tumor microenvironment, we studied the impact of vecabrutinib specifically on T-cell subsets. Firstly, no difference in the proportions of CD4 or CD8 expressing T-cells was observed in mice treated with vehicle or vecabrutinib. However, of interest, the percentage of CD4+ CD25+ FoxP3+ regulatory T cells (Tregs) were significantly decreased upon treatment with vecabrutinib in peripheral blood (P=0.026) and spleen (P=0.009). The decrease in Tregs was due to reduced proliferation of these cells upon exposure to the drug as measured by Ki-67 staining. Also, the Tregs expressing the maturation and activation markers such as CD103 and GITR were significantly decreased in blood and spleen upon drug treatment. Further, we analysed the changes in CD8 T-cell subsets following treatment with vecabrutinib. Treatment with the drug resulted in expansion of the CD127+ CD44- naïve CD8 T-cells in blood, bone marrow and spleen (all P values 0.002) while the CD127+ CD44+ memory CD8 T-cells were significantly decreased in bone marrow and spleen (all P values 0.009). Also, the CD127low CD44int-hi effector CD8 T-cells were decreased in blood (P=0.004), bone marrow (P=0.004) and spleen (P=0.002) upon vecabrutinib treatment. Therefore, vecabrutinib treatment did not alter the percentage of CD4+ and CD8+ T cells in mice however, significant changes in the subset composition of the CD4 and CD8 T cells were observed. Lastly, to analyse the impact of vecabrutinib on survival, a cohort of mice (n=12) were transplanted with 5 million splenic tumor cells isolated from Eµ-TCL1 transgenic mice. After allowing for engraftment, the mice were randomized to treatment with the drug (n=6) or vehicle (n=6). Of note, the mice treated with the drug showed a significant increase in survival (median 35 days from transplant; P In summary, vecabrutinib was efficacious in vivo in a preclinical CLL adoptive transfer model, decreasing tumor burden in different organs and significantly improving survival. Treatment with the drug altered the T-cell architecture in vivo. Of interest, the immunosuppressive Tregs, which protect the tumor from immune surveillance were decreased in various tissue compartments; however, a decrease in the effector CD8 T cells might impact anti-tumor immunity if there is a consistent effect upon drug treatment. Vecabrutinib antitumor activity and effects on T-cell populations in vivo in this preclinical CLL model are intriguing, merits further investigation and supports the ongoing phase 1b/2 study in patients with previously treated B-lymphoid malignancies (NCT03037645). Disclosures Tausch: AbbVie: Consultancy, Other: Travel grants; Celgene: Consultancy, Other: Travel grants; Gilead: Consultancy, Other: Travel grants. Fox:Sunesis Pharmaceuticals: Employment; Amphivena Therapeutics: Employment. Taverna:Sunesis Pharmaceuticals: Employment. Stilgenbauer:Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Boehringer-Ingelheim: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Hoffmann La-Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GSK: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmcyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Mundipharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
28. Phase I and Pharmacologic Study of SNS-032, a Potent and Selective Cdk2, 7, and 9 Inhibitor, in Patients With Advanced Chronic Lymphocytic Leukemia and Multiple Myeloma
- Author
-
Ashraf Badros, R. Donald Harvey, Rong Chen, Tianling Chen, Kristi Mahadocon, Judith A. Fox, Rajni Sinha, David A. Siegel, Wei Gang Tong, William G. Wierda, Peggy Kegley, William Plunkett, Ute Hoch, Steven Coutre, and Leslie Popplewell
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Chronic lymphocytic leukemia ,Loading dose ,Cohort Studies ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Tissue Distribution ,Oxazoles ,Multiple myeloma ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,Cyclin-Dependent Kinase 2 ,ORIGINAL REPORTS ,Middle Aged ,medicine.disease ,Cyclin-Dependent Kinase 9 ,Leukemia, Lymphocytic, Chronic, B-Cell ,Cyclin-Dependent Kinases ,XIAP ,Survival Rate ,Tumor lysis syndrome ,Thiazoles ,Leukemia ,Treatment Outcome ,Pharmacodynamics ,Toxicity ,Immunology ,Female ,Multiple Myeloma ,business ,Cyclin-Dependent Kinase-Activating Kinase - Abstract
Purpose SNS-032 is a highly selective and potent inhibitor of cyclin-dependent kinases (Cdks) 2, 7, and 9, with in vitro growth inhibitory effects and ability to induce apoptosis in malignant B cells. A phase I dose-escalation study of SNS-032 was conducted to evaluate safety, pharmacokinetics, biomarkers of mechanism-based pharmacodynamic (PD) activity, and clinical efficacy. Patients and Methods Parallel cohorts of previously treated patients with chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) received SNS-032 as a loading dose followed by 6-hour infusion weekly for 3 weeks of each 4-week course. Results There were 19 patients with CLL and 18 with MM treated. Tumor lysis syndrome was the dose-limiting toxicity (DLT) for CLL, the maximum-tolerated dose (MTD) was 75 mg/m2, and the most frequent grade 3 to 4 toxicity was myelosuppression. One patient with CLL had more than 50% reduction in measurable disease without improvement in hematologic parameters. Another patient with low tumor burden had stable disease for four courses. For patients with MM, no DLT was observed and MTD was not identified at up to 75 mg/m2, owing to early study closure. Two patients with MM had stable disease and one had normalization of spleen size with treatment. Biomarker analyses demonstrated mechanism-based PD activity with inhibition of Cdk7 and Cdk9, decreases in Mcl-1 and XIAP expression level, and associated CLL cell apoptosis. Conclusion SNS-032 demonstrated mechanism-based target modulation and limited clinical activity in heavily pretreated patients with CLL and MM. Further single-agent, PD-based, dose and schedule modification is warranted to maximize clinical efficacy.
- Published
- 2010
- Full Text
- View/download PDF
29. Voreloxin, a first-in-class anticancer quinolone derivative, acts synergistically with cytarabine in vitro and induces bone marrow aplasia in vivo
- Author
-
Jeffrey A. Silverman, Jeffrey L. Kumer, Anthony Howlett, Caroline Darne Scatena, Jennifer P. Arbitrario, Rachael E. Hawtin, and Judith A. Fox
- Subjects
Blood Platelets ,Cancer Research ,medicine.drug_class ,DNA damage ,Bone Marrow Cells ,HL-60 Cells ,Bone Marrow Aplasia ,Anthracycline ,Pharmacology ,Toxicology ,Vosaroxin ,Mice ,chemistry.chemical_compound ,AML ,Leukemia, Promyelocytic, Acute ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Leukocytes ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Naphthyridines ,Voreloxin ,Cell Proliferation ,biology ,Topoisomerase ,Cytarabine ,Myeloid leukemia ,Drug Synergism ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Bone marrow ablation ,Quinolone ,medicine.disease ,Topoisomerase II ,Disease Models, Animal ,Thiazoles ,Leukemia ,Oncology ,chemistry ,biology.protein ,Original Article ,Female ,medicine.drug - Abstract
Main purpose Voreloxin is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, inducing site-selective DNA damage. Voreloxin is in clinical studies, as a single agent and in combination with cytarabine, for the treatment of acute myeloid leukemia (AML). The preclinical studies reported here were performed to investigate the activity of voreloxin alone and in combination with cytarabine, in support of the clinical program. Research questions Is single agent voreloxin active in preclinical models of AML? Does the combination of voreloxin and cytarabine enhance the activity of either agent alone? Methods Inhibition of proliferation was studied in three cancer cell lines: HL-60 (acute promyelocytic leukemia), MV4-11 (AML), and CCRF-CEM (Acute lymphoblastic leukemia). Combination index (CI) analysis established the effect of the drugs in combination. A mouse model of bone marrow ablation was used to investigate in vivo efficacy of the drugs alone and in combination. Peripheral white blood cell and platelet counts were followed to assess marrow impact and recovery. Results Voreloxin and cytarabine alone and in combination exhibited cytotoxic activity in human leukemia cell lines and in vivo. The two drugs had additive or synergistic activity in vitro and supra-additive activity in vivo. Bone marrow ablation was accompanied by reductions in peripheral white blood cells and platelets that were reversible within 1 week, consistent with the AML treatment paradigm. Conclusions These data support ongoing clinical evaluation of voreloxin both alone and in combination with cytarabine for the treatment of AML.
- Published
- 2010
- Full Text
- View/download PDF
30. A Phase I Pharmacokinetic and Pharmacodynamic Study of TKI258, an Oral, Multitargeted Receptor Tyrosine Kinase Inhibitor in Patients with Advanced Solid Tumors
- Author
-
R. Morrison, Edgar Braendle, Carla Heise, Felix Garzon, Priska Butzberger-Zimmerli, Judith A. Fox, Debashis Sarker, Dalal Jadayel, T.R. Jeffrey Evans, R. Molife, Johann S. de Bono, Sharianne Louie, C. Marriott, Natasha Aziz, Ian Judson, Maryon Hardie, and Glenn C. Michelson
- Subjects
Adult ,Male ,Cancer Research ,Maximum Tolerated Dose ,Nausea ,Administration, Oral ,Antineoplastic Agents ,Quinolones ,Pharmacology ,Pharmacokinetics ,Oral administration ,Neoplasms ,Humans ,Medicine ,Dosing ,Protein Kinase Inhibitors ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Middle Aged ,Protein-Tyrosine Kinases ,Treatment Outcome ,Oncology ,Pharmacodynamics ,Toxicity ,Vomiting ,Benzimidazoles ,Female ,medicine.symptom ,business ,Off Treatment - Abstract
Purpose: To determine the maximum tolerated dose (MTD) dose-limiting toxicity, and pharmacokinetic and pharmacodynamic profile of TKI258 (formerly CHIR-258). Experimental Design: A phase I dose escalating trial in patients with advanced solid tumors was performed. Treatment was initially as single daily doses on an intermittent 7-day on/7-day off schedule. Following a protocol amendment, a second schedule comprised, during cycle 1, 7-day on/7-day off treatment followed by 14 days of continuous daily dosing; subsequent cycles comprised 28 days of daily dosing. Pharmacokinetics and evaluation of phosphorylated extracellular signal-regulated kinase (ERK) in peripheral blood mononuclear cells were done during the first 28 days of each schedule. Results: Thirty-five patients were treated in four intermittent (25-100 mg/d) and three continuous (100-175 mg/d) dosing cohorts. Observed drug-related toxicities were nausea and vomiting, fatigue, headache, anorexia, and diarrhea. Dose-limiting toxicities were grade 3 hypertension in one patient at 100 mg continuous dosing, grade 3 anorexia in a second patient at 175 mg, and grade 3 alkaline phosphatase elevation in a third patient at 175 mg. One patient had a partial response (melanoma) and two patients had stable disease >6 months. TKI258 pharmacokinetics were linear over the dose range of 25 to 175 mg. Five of 14 evaluable patients had modulation of phosphorylated ERK levels. Conclusions: The MTD was defined as 125 mg/d. Evidence of antitumor activity in melanoma and gastrointestinal stromal tumors warrants further investigation, and other phase I studies are ongoing. Further pharmacodynamic evaluation is required in these studies to evaluate the biological effects of TKI258.
- Published
- 2008
- Full Text
- View/download PDF
31. Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia
- Author
-
Michael Weichel, Chelsea J. Gudgeon, Kristina Ellwanger, Uwe Reusch, Stefan Knackmuss, Jeanmarie Guenot, Ivica Fucek, Roland B. Walter, Kimberly H. Harrington, Judith A. Fox, Eugene A. Zhukovsky, and Lori Kunkel
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,CD3 Complex ,Gemtuzumab ozogamicin ,medicine.medical_treatment ,T-Lymphocytes ,CD33 ,Sialic Acid Binding Ig-like Lectin 3 ,Biology ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Avidity ,Binding Sites ,Myeloid leukemia ,Antibodies, Monoclonal ,Immunotherapy ,medicine.disease ,Gemtuzumab ,Leukemia ,Cytolysis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Aminoglycosides ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine.drug ,Half-Life - Abstract
Purpose: Randomized studies with gemtuzumab ozogamicin have validated CD33 as a target for antigen-specific immunotherapy of acute myelogenous leukemia (AML). Here, we investigated the potential of CD33/CD3-directed tandem diabodies (TandAbs) as novel treatment approach for AML. These tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a bivalent antibody and have a molecular weight exceeding the renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs. Experimental Design: We constructed a series of TandAbs composed of anti-CD33 and anti-CD3 variable domains of diverse binding affinities and profiled their functional properties in CD33+ human leukemia cell lines, xenograft models, and AML patient samples. Results: Our studies demonstrated that several CD33/CD3 TandAbs could induce potent, dose-dependent cytolysis of CD33+ AML cell lines. This effect was modulated by the effector-to-target cell ratio and strictly required the presence of T cells. Activation and proliferation of T cells and maximal AML cell cytolysis correlated with high avidity to both CD33 and CD3. High-avidity TandAbs were broadly active in primary specimens from patients with newly diagnosed or relapsed/refractory AML in vitro, with cytotoxic properties independent of CD33 receptor density and cytogenetic risk. Tumor growth delay and inhibition were observed in both prophylactic and established HL-60 xenograft models in immunodeficient mice. Conclusions: Our data show high efficacy of CD33/CD3 TandAbs in various preclinical models of human AML. Together, these findings support further study of CD33/CD3 TandAbs as novel immunotherapeutics for patients with AML. Clin Cancer Res; 22(23); 5829–38. ©2016 AACR.
- Published
- 2016
32. Characterization of a CC49-Based Single-Chain Fragment−β-Lactamase Fusion Protein for Antibody-Directed Enzyme Prodrug Therapy (ADEPT)
- Author
-
Joshua Roy Basler, Lilia Babé, Peter D. Senter, Volker Schellenberger, Enrique Escandon, Ralph F. Alderson, Martin Roberge, Wei Geng, Brian E. Toki, Regina Chin, Roanna Ueda, Judith A. Fox, Tianling Chen, Amy Liu, Tessi Kanavarioti, and Douglas Hodges
- Subjects
Antibodies, Neoplasm ,Recombinant Fusion Proteins ,Cell ,Immunoglobulin Variable Region ,Biomedical Engineering ,Mice, Nude ,Pharmaceutical Science ,Mutagenesis (molecular biology technique) ,Bioengineering ,Irinotecan ,beta-Lactamases ,Epitope ,Mice ,Drug Delivery Systems ,medicine ,Animals ,Humans ,Prodrugs ,Antineoplastic Agents, Alkylating ,Immunoglobulin Fragments ,Melphalan ,Binding selectivity ,Pharmacology ,Drug Carriers ,Antibiotics, Antineoplastic ,Molecular Structure ,biology ,Chemistry ,Organic Chemistry ,Adept ,Prodrug ,Antineoplastic Agents, Phytogenic ,Fusion protein ,Molecular biology ,Cephalosporins ,medicine.anatomical_structure ,Biochemistry ,Doxorubicin ,Nitrogen Mustard Compounds ,biology.protein ,Camptothecin ,Female ,Antibody ,Colorectal Neoplasms ,Neoplasm Transplantation ,Biotechnology - Abstract
CC49 is a clinically validated antibody with specificity for TAG-72, a carbohydrate epitope that is overexpressed and exposed on the cell surface in a large fraction of solid malignancies. We constructed a single-chain fragment (scFv) based on CC49 and fused it to beta-lactamase (BLA). Following optimization of the scFv domain by combinatorial consensus mutagenesis (CCM) for increased expression and stability, we characterized the protein variant for binding, in vivo pharmacokinetics (PK), and antitumor efficacy. The fusion protein TAB2.5 possessed a similar binding specificity relative to the parent antibody CC49. TAB2.5 also showed prolonged retention (T(1/2) = 36.9 h) in tumor-bearing mice with tumor/plasma ratios of up to 1000. Preliminary evaluation of TAB2.5, in combination with a novel prodrug, GC-Mel, resulted in significant efficacy in a colorectal xenograft tumor model and supports the utility of the protein as an agent for tumor-selective prodrug activation.
- Published
- 2006
- Full Text
- View/download PDF
33. TISSUE DISTRIBUTION AND RECEPTOR-MEDIATED CLEARANCE OF ANTI-CD11A ANTIBODY IN MICE
- Author
-
Anahid Bakshi, Judith A. Fox, Susanne Pippig, Paul J. Fielder, Michelle Gonzales, Barbara Reitz, Josette Padilla-Eagar, Susan Palmieri, and Greg P. Coffey
- Subjects
Male ,Neutrophils ,Lymphocyte ,media_common.quotation_subject ,Efalizumab ,Pharmaceutical Science ,CD11a ,Biology ,Antibodies, Monoclonal, Humanized ,Peripheral blood mononuclear cell ,Mice ,Microscopy, Electron, Transmission ,Antigen ,Bone Marrow ,In vivo ,medicine ,Animals ,Tissue Distribution ,CD11a Antigen ,Internalization ,media_common ,Pharmacology ,Macrophages ,Antibodies, Monoclonal ,hemic and immune systems ,Flow Cytometry ,Immunohistochemistry ,Molecular biology ,Receptors, Antigen ,medicine.anatomical_structure ,Liver ,Immunology ,biology.protein ,Autoradiography ,Lymph Nodes ,Antibody ,Spleen ,Subcellular Fractions ,medicine.drug - Abstract
Efalizumab (Raptiva) is a humanized monoclonal antibody specific for CD11a, the alpha-chain component of the lymphocyte function-associated antigen 1. In humans, the rate of efalizumab elimination from serum was related to the level of CD11a cell surface expression. These data suggested a role for the CD11a receptor, itself, in efalizumab clearance. Recently, we conducted a series of in vitro studies that suggested a role for CD11a-expressing T cells in efalizumab clearance as mediated by cellular internalization and lysosome-mediated degradation (Coffey et al., 2004). To further study the mechanism of anti-CD11a clearance in vivo, we assessed the tissue distribution, cellular internalization, and subcellular localization of a rat anti-mouse CD11a monoclonal antibody in various tissues in mice. Anti-CD11a antibody primarily distributed to leukocytes and macrophages in the peripheral blood, spleen, and liver, with uptake in the lymph nodes and bone marrow after 72 h. At least a portion of the antibody was internalized and cleared by peripheral blood mononuclear cells, lymphocytes, and splenocytes in a time-dependent manner in vivo. Internalized antibody costained with LysoTracker Red, suggesting that it was transported to lysosomes for degradation. Together, these data suggest that one clearance mechanism for anti-CD11a antibody in vivo is via receptor-mediated internalization and lysosomal degradation by CD11a-expressing cells and tissues.
- Published
- 2005
- Full Text
- View/download PDF
34. How to Kill a Zombie: Strategies for Dealing with the Aftermath of the Foreclosure Crisis
- Author
-
Judith L. Fox
- Subjects
Order (exchange) ,Law ,Political economy ,Zombie ,Control (management) ,Economics ,Legislature ,Foreclosure ,Loss mitigation - Abstract
The foreclosure crisis which began in 2008 is old news; or is it? A lot of attention has been paid to the plight of homeowners struggling to save their homes from foreclosure. Legislative and regulatory changes have made it easier for homeowners to navigate the loss mitigation process. A significant number of people, however, did not try to save their homes. In fact, some actively tried unsuccessfully to give the homes back to their lender. These abandoned homes and abandoned foreclosures have become zombie mortgages. This is the legacy of this crisis.The existence of these homes is well documented and this paper does not seek to prove the problem. Instead, it analyzes some solutions. How can homeowners re-gain control of these homes in order to solve the urban plight problem the foreclosure crisis left in its wake? Current law does not anticipate a bank not seeking foreclosure. Should it? Courts are just beginning to grapple with the situation. No consistent patterns have emerged to deal with the issues. The mortgage industry has argued throughout this crisis that delays in judicial foreclosure are one of the causes of this phenomena. The data has failed to support this hypothesis. On the contrary, this paper argues that the industry used every legal maneuver possible to exacerbate the problem. Lenders have been allowed to enjoy the benefits of the mortgage, while avoiding the burdens. Changes in policy and perception are needed if we are ever to move pass the crisis.
- Published
- 2015
- Full Text
- View/download PDF
35. Enhanced tumor killing by Apo2L/TRAIL and CPT-11 co-treatment is associated with p21 cleavage and differential regulation of Apo2L/TRAIL ligand and its receptors
- Author
-
Judith A. Fox, John B. Lowe, Mina Aikawa, Enrique Escandon, Kelly DuPree, Klara Totpal, Dominick Sinicropi, and Hong Xiang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Receptor expression ,Apoptosis ,Biology ,Irinotecan ,medicine.disease_cause ,Receptors, Tumor Necrosis Factor ,TNF-Related Apoptosis-Inducing Ligand ,Downregulation and upregulation ,Cyclins ,Neoplasms ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,RNA, Neoplasm ,Decoy receptors ,Receptor ,Molecular Biology ,Cells, Cultured ,Membrane Glycoproteins ,Tumor Necrosis Factor-alpha ,Kinase ,Carcinoma ,Cell Cycle ,Drug Synergism ,Cell cycle ,Antineoplastic Agents, Phytogenic ,Kinetics ,Biochemistry ,Caspases ,Colonic Neoplasms ,Cancer research ,Camptothecin ,Apoptosis Regulatory Proteins ,Carcinogenesis - Abstract
Apo2L/TRAIL exhibits enhanced apoptotic activity in tumor xenograft models when used in combination with the topoisomerase 1 inhibitor CPT-11. To investigate the cellular mechanisms involved in this increased tumor-killing activity, a series of in vitro experiments were conducted using the human colon carcinoma cell line (HCT116). Apo2L/TRAIL induced a transient upregulation of DR5 mRNA, while CPT-11 increased both death and decoy receptor expression. Upregulation of decoy receptors by CPT-11 was partially inhibited by co-administration of Apo2L/TRAIL. CPT-11 treatment resulted in accumulation of cells at G(2)M-phase and correlated with a substantial increase in the protein levels of the cyclin-dependent kinase inhibitor p21. However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. The sequential treatment produced the greatest induction of DR5 and DR4, caspase-3-like cleavage/activation and p21 degradation, as well as increased apoptosis. These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis.
- Published
- 2002
- Full Text
- View/download PDF
36. Pharmacokinetics of rhuMAb CD18, a Recombinant Humanised Monoclonal Antibody Fragment to CD18, in Normal Healthy Human Volunteers
- Author
-
Judith A. Fox, Steven G. Gourlay, Eugene Koren, Robert M. Miller, and David E. Allison
- Subjects
Adult ,Male ,Pharmacology ,Placebo ,Immunoglobulin Fab Fragments ,Bolus (medicine) ,Pharmacokinetics ,White blood cell ,Humans ,Medicine ,Single-Blind Method ,Pharmacology (medical) ,Adverse effect ,Volume of distribution ,biology ,business.industry ,Antibodies, Monoclonal ,General Medicine ,Middle Aged ,Recombinant Proteins ,medicine.anatomical_structure ,Tolerability ,Area Under Curve ,CD18 Antigens ,Antibody Formation ,Injections, Intravenous ,biology.protein ,Female ,Antibody ,business ,Biotechnology - Abstract
Background and Objectives: Leucocyte β2 integrin adhesion receptors are hypothesised as a therapeutic target to modify immune responses to ischaemia-reperfusion injury that may be detrimental to recovery in a variety of disease states. Two phase I studies were designed to evaluate the pharmacokinetics, immunogenicity and safety of rhuMAb CD18, ahumanised monoclonal antibody F(ab’)2 fragment to the CD18 receptor, in normal healthy human volunteers. Study Design and Methods: The first study evaluated six escalating doses of rhuMAb CD18 (0.06, 0.12, 0.25, 0.5, 1.0, 2.0 mg/kg) in 36 subjects given two intravenous (IV) bolus injections 12 hours apart. In the second study, 16 subjects received IV doses of 1.0 and 2.0 mg/kg as a single dose or as two doses given 12 hours apart. Study endpoints were rhuMAb CD18 serum pharmacokinetics, change in white blood cell (WBC) count, and safety and tolerability. The two studies enrolled a total of 53 subjects. Results: Serum concentration-time profiles demonstrated a monophasic decline and were best characterised by a one-compartment pharmacokinetic model. At the doses administered, the volume of distribution approximated the serum volume (range of means: 42 to 58 ml/kg). The serum clearance decreased with increasing dose until becoming consistent at doses of 0.5 to 2.0 mg/kg (range of means: 3.1 to 5.0 ml/h/kg). At doses of 0.5 to 2.0 mg/kg, the mean elimination half-life ranged from 7.0 to 9.6 hours. WBC counts increased at doses of above 0.06 mg/kg, returning to within 20% of predose values by day 7. Antibodies to rhuMAb CD18 were not detected at day 28. Mild-to-moderate adverse events were observed in both the placebo and treated groups, and were limited to flu-like symptoms. One subject experienced a serious adverse event (febrile reaction) and recovered with minimal intervention. There was no evidence of an increase in infection in subjects who received rhuMAb CD18. Conclusions: Upon IV bolus administration, rhuMAb CD18 serum concentration-time data fit a one-compartment pharmacokinetic model. At doses of 0.5 to 2.0 mg/kg, the pharmacokinetics were linear and the half-life ranged from 7.0 to 9.6 hours with a volume of distribution that approximated the serum volume. No antibodies to rhuMAb CD18 were detected. A transient, dose-dependent increase in the WBC count was observed, consistent with the expected effect of rhuMAb CD18 on leucocyte demargination. No increase in infection was observed. rhuMAb CD18 administered by IV bolus was well tolerated, with the exception of one febrile reaction.
- Published
- 2002
- Full Text
- View/download PDF
37. Abstract 1207: SNS-062 demonstrates efficacy in chronic lymphocytic leukemia in vitro and inhibits C481S mutated Bruton tyrosine kinase
- Author
-
Amy J. Johnson, Judith A. Fox, Linda L. Neuman, John C. Byrd, Daphne Guinn, Jennifer A. Woyach, Catherine A. Fabian, Sean D. Reiff, and Wendy Wilson
- Subjects
Cancer Research ,animal structures ,T cell ,Jurkat cells ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Medicine ,Bruton's tyrosine kinase ,Viability assay ,Kinase activity ,biology ,business.industry ,CD28 ,Molecular biology ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,Immunology ,biology.protein ,Acalabrutinib ,business ,030215 immunology - Abstract
Introduction: In order to address the issue of acquired resistance to ibrutinib, we sought to characterize the Bruton agammaglobulinemia tyrosine kinase (BTK) inhibitor SNS-062 in preclinical models of chronic lymphocytic leukemia (CLL). Methods: Primary CLL B cells were isolated from the whole blood of consented patients by ficoll density centrifugation and Rosette-Sep negative selection. Annexin V and propidium iodide flow cytometry was used to measure patient CLL cell viability and 7-AAD was used to measure viability in stromal co-culture. CD40 and CD86 expression was evaluated via flow cytometry subsequent to sustained 3.2uM CpG stimulation. BCR signaling in primary CLL cells was investigated by immunoblot following 1 hour treatment and following 1 hour or 24 hours of incubation with SNS-062 in XLA cell lines. ITK inhibition was investigated via immunoblot after stimulation with anti-CD3 and anti-CD28 and incubation with SNS-062 for 1 hour. SNS-062 was used at a concentration of 1uM in preclinical studies unless otherwise noted. Measurement of kinase activity in human recombinant WT BTK or C481S BTK was performed in a FRET kinase assay. Results: Immunoblots of BTK and ERK phosphorylation of XLA cells transfected with WT or C481S BTK demonstrated that SNS-062 inhibition is comparable to that of ibrutinib in WT BTK and greater than that of ibrutinib in C481S BTK. Using a recombinant kinase assay, we found the IC50 of SNS-062 against WT BTK to be 4.6nM and C481S BTK to be 1.1nM, suggesting that SNS-062 retains activity against the mutated BTK variant. Additionally, SNS-062 was found to be six times more potent than ibrutinib and greater than 640 times more potent than acalabrutinib against C481S BTK. SNS-062 demonstrates dose-dependent inhibition of BTK in primary patient CLL cells comparable to ibrutinib via immunoblot for BTK phosphorylation. The viability of primary patient cells treated with 0.1uM, 1.0uM, and 10.0uM SNS-062 for 48 hours was measured to be 96.7%, 96.1%, and 88.1%, respectively, that of the untreated condition. At 48 hours, SNS-062 decreased viability of primary CLL cells in the presence of HS5 stromal protection by 5.5%. SNS-062 was found to decrease CpG induced CD40 and CD86 expression by 8.7% and 15.7%, respectively. Using an in vitro kinase assay, SNS-062 inhibited ITK with an IC50 value of 24nM. An immunoblot of anti-CD3/CD28 stimulated Jurkat cells revealed that SNS-062 decreased the phosphorylation of ERK, implying inhibition of ITK. Conclusion: Unlike ibrutinib, SNS-062 inhibits BTK signaling in the presence of the C481S mutation and may address acquired resistance to covalent BTK inhibitors. SNS-062 decreases B cell activation markers, viability, and stromal cell protection in primary patient CLL cells and was shown to inhibit ITK, suggesting support of T cell mediated antitumor activities. These data support further investigation of this molecule and advancement into clinical trials. Citation Format: Catherine A. Fabian, Sean D. Reiff, Daphne Guinn, Linda Neuman, Judith A. Fox, Wendy Wilson, John C. Byrd, Jennifer A. Woyach, Amy J. Johnson. SNS-062 demonstrates efficacy in chronic lymphocytic leukemia in vitro and inhibits C481S mutated Bruton tyrosine kinase [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 1207. doi:10.1158/1538-7445.AM2017-1207
- Published
- 2017
- Full Text
- View/download PDF
38. High Resolution Mapping of the Binding Site on Human IgG1 for FcγRI, FcγRII, FcγRIII, and FcRn and Design of IgG1 Variants with Improved Binding to the FcγR
- Author
-
Josephine P. Briggs, Kyu Hong, Leonard G. Presta, Betty Li, Angela K. Namenuk, Judith A. Fox, Dong Xie, Julie Rae, Y. Gloria Meng, Jadine Lai, Andrew Stadlen, and Robert L. Shields
- Subjects
Glycosylation ,biology ,Effector ,Cell Biology ,Protein engineering ,Biochemistry ,Molecular biology ,Immunoglobulin G ,Cell biology ,chemistry.chemical_compound ,Neonatal Fc receptor ,chemistry ,biology.protein ,Binding site ,Antibody ,Receptor ,Molecular Biology - Abstract
Immunoglobulin G (IgG) Fc receptors play a critical role in linking IgG antibody-mediated immune responses with cellular effector functions. A high resolution map of the binding site on human IgG1 for human FcγRI, FcγRIIA, FcγRIIB, FcγRIIIA, and FcRn receptors has been determined. A common set of IgG1 residues is involved in binding to all FcγR; FcγRII and FcγRIII also utilize residues outside this common set. In addition to residues which, when altered, abrogated binding to one or more of the receptors, several residues were found that improved binding only to specific receptors or simultaneously improved binding to one type of receptor and reduced binding to another type. Select IgG1 variants with improved binding to FcγRIIIA exhibited up to 100%enhancement in antibody-dependent cell cytotoxicity using human effector cells; these variants included changes at residues not found at the binding interface in the IgG/FcγRIIIA co-crystal structure (Sondermann, P., Huber, R., Oosthuizen, V., and Jacob, U. (2000)Nature 406, 267–273). These engineered antibodies may have important implications for improving antibody therapeutic efficacy.
- Published
- 2001
- Full Text
- View/download PDF
39. Immunotherapy approach to allergic disease
- Author
-
Judith A. Fox, Paula M. Jardieu, and Robert B. Fick
- Subjects
Allergy ,medicine.drug_class ,medicine.medical_treatment ,medicine.disease_cause ,Monoclonal antibody ,Immunoglobulin E ,Immune complex formation ,Epitope ,Hypersensitivity ,Animals ,Humans ,Medicine ,Pharmacology ,biology ,business.industry ,Antibodies, Monoclonal ,Immunotherapy ,Allergens ,medicine.disease ,Immunology ,Allergic response ,biology.protein ,Antibody ,business - Abstract
The causal role of immunoglobulin E (IgE) in triggering the cascade of biochemical events leading to allergic disease is well established. Treatments that selectively inhibit IgE activity are a logical approach in managing the allergic response. One such strategy utilizes rhuMAb-E25, a recombinant humanized IgG 1 monoclonal anti-IgE antibody, which binds to IgE. This anti-IgE antibody binds at the same epitope site of IgE that binds to FceRI and is thus non-anaphylactogenic. By binding to IgE and removing it via immune complex formation, the pool of IgE available to interact with mast cells and basophils is thereby reduced and the allergic response is attenuated. The clinical safety and efficacy of rhuMAb-E25 demonstrated in phase II studies of allergic asthma will be outlined.
- Published
- 2000
- Full Text
- View/download PDF
40. Secretion of glycosylation site mutants can be rescued by the signal/pro sequence of tissue plasminogen activator
- Author
-
Dave Brousseau, Judith A. Fox, Adriana Johnson, David H. Peers, Phillip W. Berman, Thomas Ryll, Sabrina Tom, Timothy A. Hotaling, Richard L. Gehant, Steven M. Chamow, and Christiane Köhne
- Subjects
Signal peptide ,chemistry.chemical_classification ,Glycosylation ,Endoplasmic reticulum ,Cell Biology ,Golgi apparatus ,Biology ,Biochemistry ,carbohydrates (lipids) ,chemistry.chemical_compound ,symbols.namesake ,chemistry ,symbols ,Secretion ,Binding site ,Glycoprotein ,Molecular Biology ,Peptide sequence - Abstract
Strategies that prevent the attachment of N-linked carbohydrates to nascent glycoproteins often impair intracellular transport and secretion. In the present study, we describe a method to rescue the intracellular transport and secretion of glycoproteins mutagenized to delete N-linked glycosylation sites. Site-directed mutagenesis was used to delete N-linked glycosylation sites from a chimeric protein, TNFR-IgG1. Deletion of any of the three glycosylation sites in the TNFR portion of the molecule, alone or in combination, resulted in a moderate or near total blockade of TNFR-IgG1 intracellular transport and secretion. Pulse chase experiments suggested that the glycosylation site mutants accumulated in the endoplasmic reticulum (ER) and were inefficiently exported to the Golgi apparatus (GA). Replacement of the TNFR signal sequence with the signal/pro sequence of human tissue plasminogen activator (tPA) overcame the blockade to intracellular transport, and restored secretion to levels comparable to those achieved with the fully glycosylated molecule. Ligand binding studies suggested that the secreted glycosylation variants possessed binding characteristics similar to the fully glycosylated protein. This study demonstrates that N-terminal sequences of tPA are unexpectedly efficient in facilitating transport from the ER to the GA and suggests that these sequences contain a previously unrecognized structural element that promotes intracellular transport. J. Cell. Biochem. 75:446–461, 1999. © 1999 Wiley-Liss, Inc.
- Published
- 1999
- Full Text
- View/download PDF
41. Expanded Close-Packed Fullerides: The Reactivity of Na2C60 with Ammonia
- Author
-
Paul F. Henry, Stephen J. Heyes, Matthew J. Rosseinsky, Amelia J. Fowkes, and Judith M. Fox
- Subjects
Ammonia ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Chemistry ,Inorganic chemistry ,Reactivity (chemistry) ,General Chemistry ,Trigonal crystal system ,Biochemistry ,Catalysis - Abstract
A novel expanded fulleride results from the room temperature reaction of Na2C60 with ammonia. The rhombohedral packing of (ND3)8Na2C60 arises from cooperative rearrangement of the face-centered cub...
- Published
- 1997
- Full Text
- View/download PDF
42. New Metal Fullerides
- Author
-
Judith M. Fox, A. J. Fowkes, K. M. Allen, A. C. Duggan, Matthew J. Rosseinsky, and Paul F. Henry
- Subjects
Metal ,Fullerene ,Materials science ,Chemical physics ,General Chemical Engineering ,visual_art ,Interstitial defect ,visual_art.visual_art_medium ,Knight shift ,Electronic structure ,Magnetic susceptibility ,Ammonium compounds - Abstract
The article is a summary of recent work on the synthesis of new types of metal fulleride designed to examine the influence of various chemical factors on the physical properties of this class of material. Fullende charge spacing, orientations are examined, together with the insertion of less electropositive cations on the interstitial sites.
- Published
- 1997
- Full Text
- View/download PDF
43. First-in-Human Phase 1a Study of the Safety, Pharmacokinetics, and Pharmacodynamics of the Noncovalent Bruton Tyrosine Kinase (BTK) Inhibitor SNS-062 in Healthy Subjects
- Author
-
Deena Gruver, Renee Ward, Langdon L. Miller, David Arnold, Linda L. Neuman, Josué Mfopou Kunjom, Judith A. Fox, Wendy Hill, and Daniel L. Combs
- Subjects
0301 basic medicine ,business.industry ,Nausea ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,Placebo ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Ibrutinib ,Pharmacodynamics ,Clinical endpoint ,Medicine ,Acalabrutinib ,medicine.symptom ,business ,Adverse effect - Abstract
Background: SNS-062 is a potent, noncovalent (reversible) BTK inhibitor in development for B-cell malignancies and other cancers. SNS-062 has the potential for activity in patients whose cancers are sensitive to BTK inhibition, as well as those that are resistant to ibrutinib through acquisition of a BTK Cys481Ser mutation. In vitro studies have demonstrated that SNS-062 antitumor activity in cells with the mutation is unaffected (Binnerts et al, EORTC 2015, Abstract C186), in contrast to the substantially reduced activity seen with ibrutinib and acalabrutinib. This study was designed to evaluate the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of SNS-062 in healthy subjects. Methods: This was a phase 1a, first-in-human, randomized, double-blind, placebo-controlled, sequential-group, single-dose study conducted in 3 stages. In stage 1, four sequential cohorts of 8 subjects were randomly assigned to receive ascending SNS-062 dose levels (50, 100, 200, and 300 mg; n=6, 3 male, 3 female) or placebo (n=2, 1 male, 1 female) as a single dose administered orally. The primary endpoint was safety, assessed by adverse events (AEs), laboratory parameters, and cardiac monitoring. Secondary endpoints included PK parameters and PD parameters (inhibition of phosphorylation BTK [pBTK] as determined by ELISA in whole blood lysates). Stages 2 and 3 were designed to evaluate the effects of food and CYP3A4 inhibition, respectively, on the PK of SNS-062. Results: In stage 1 (n=32), the median age was 55 years (range: 22-64) among those who received SNS-062 (n=24) and 42.5 years (range: 29-65) among those who received placebo (n=8). Treatment-emergent AEs (TEAEs) were reported for 8 (33%) subjects who received SNS-062 and for 3 (38%) subjects who received placebo. TEAEs reported for subjects who received SNS-062 included headache (n=5) and nausea, constipation, bronchitis, fatigue, orthostatic hypotension, and supraventricular tachycardia (n=1 each) without obvious evidence of dose dependency. AEs in the placebo group included headache (n=2), nausea (n=2), and diarrhea (n=1). AEs were all reported as Grade 1 except for 1 subject (who received 300 mg SNS-062) who experienced Grade 2 headache and fatigue. No Grade 3 or higher AEs and no serious AEs were reported. SNS-062 was rapidly absorbed (median Tmax: 1 hour [range: 0.5-3.0 hours]). SNS-062 concentrations declined in a multiphasic manner. Exposure increased approximately proportional to dose. Mean PK parameters for each cohort are shown in the Table. SNS-062 demonstrated rapid and near complete inhibition of pBTK at all dose levels. Stages 2 and 3 are in progress and results of the completed study will be reported at the meeting. Conclusions: The observed safety, PK, and PD profiles of SNS-062 in this phase 1a study in healthy subjects support further clinical investigation. This study continues to evaluate the effects of food and CYP3A4 inhibition on SNS-062 PK. Mean SNS-062 exposure at 50 mg, the lowest dose level studied, exceeded those reported for ibrutinib (Imbruvica [package insert]. Sunnyvale, CA: Pharmacyclics, LLC; 2016) and acalabrutinib (Byrd et al, N Engl J Med 2016;374:323:32) when those drugs are administered at recommended dose levels. The extent of SNS-062 exposure and duration of pBTK inhibition are encouraging and support twice-daily dosing in a planned phase 1b/2 study in patients with advanced B-cell malignancies with and without the BTK Cys481-Ser mutation. This study was sponsored by Sunesis Pharmaceuticals. Disclosures Neuman: Puma Biotechnology: Employment; Sunesis Pharmaceuticals: Employment. Ward:Sunesis Pharmaceuticals: Consultancy, Employment. Arnold:Sunesis Pharmaceuticals: Consultancy. Combs:Sunesis Pharmaceuticals: Consultancy. Gruver:Sunesis Pharmaceuticals: Employment. Hill:Sunesis Pharmaceuticals: Employment. Miller:Sunesis Pharmaceuticals: Consultancy. Fox:Amphivena Therapeutics: Consultancy, Equity Ownership, Patents & Royalties: Patent #9212225; Bispecific CD33 and CD3 Binding Proteins; Sunesis Pharmaceuticals: Consultancy, Equity Ownership, Patents & Royalties: Patent Application #20150202189; Methods of Using SNS-595 for Treatment of Cancer Subjects with Reduced BRCA2 Activity.
- Published
- 2016
- Full Text
- View/download PDF
44. Enhancement of radiosensitivity by the novel anticancer quinolone derivative vosaroxin in preclinical glioblastoma models
- Author
-
Francesco Marampon, Luca Ventura, G.L. Gravina, Flora Vitale, Giulia Rossi, Alessandro Colapietro, Claudio Festuccia, E. Di Cesare, Andrea Mancini, and Judith A. Fox
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,medicine.drug_class ,medicine ,Radiosensitivity ,Pharmacology ,Quinolone ,medicine.disease ,Vosaroxin ,Derivative (chemistry) ,Glioblastoma - Published
- 2016
- Full Text
- View/download PDF
45. Molecular Structure of the Fulleride Anions in Superconducting K3C60 and Insulating K6C60 Determined by Powder Neutron Diffraction
- Author
-
William I. F. David, Judith M. Fox, Richard M. Ibberson, K. M. Allen, and Matthew J. Rosseinsky
- Subjects
Neutron powder diffraction ,Superconductivity ,Crystallography ,Fullerene ,Chemistry ,Ab initio quantum chemistry methods ,General Chemical Engineering ,Neutron diffraction ,Materials Chemistry ,Molecule ,General Chemistry ,Crystal structure - Abstract
The authors report here the molecular structure of K{sub 3}C{sub 60} and K{sub 6}C{sub 60} as determined by neutron powder diffraction. Structural features observed are in agreement with ab initio calculations.
- Published
- 1995
- Full Text
- View/download PDF
46. Post-partum depression: a comprehensive approach to evaluation and treatment
- Author
-
Kym Spring, Thompson and Judith E, Fox
- Subjects
Article - Abstract
Post‐partum depression (PPD) presents a significant disruption to the mother–infant relationship. Such disruptions are associated with risks to the neurological, socio‐emotional and cognitive functioning of the developing infant. A review of this literature supports the early detection of PPD and the application of comprehensive, psychotherapeutic interventions that target the functioning of the infant, the mother and the mother–infant relationship. Ecological factors important to evaluation and avenues of intervention are emphasised. The need for further research to determine evidence‐based methods of intervention is described.
- Published
- 2012
47. Radical-induced 1,3-rearrangements of allylic sulfones bearing an alkylthio or arylthio substituent at the α-position
- Author
-
Clare M. Morris, G. Darren Smyth, Judith M. Fox, and Gordon H. Whitham
- Subjects
Allylic rearrangement ,chemistry.chemical_compound ,Chemistry ,Stereochemistry ,Radical ,Substituent - Abstract
1-Methylthio-1-p-tolylsulfonylprop-2-ene 2 underwent 1,3-rearrangement under conditions of radical initiation with migration of the p-tolylsulfonyl group by a process considered to have involved addition–elimination of arylsulfonyl radicals. In contrast, the reaction of 1-p-tolylthio-1-p-tolylsulfonylprop-2-ene 3 under similar conditions gave scrambled products, indicating that arylthio and arylsulfonyl radicals participate with about equal efficiency.
- Published
- 1994
- Full Text
- View/download PDF
48. ChemInform Abstract: Na2+xHgyC60: Post-Transition Metal Intercalation Chemistry of a C60 Host
- Author
-
Judith M. Fox, Paul F. Henry, and Matthew J. Rosseinsky
- Subjects
Fullerene ,Chemistry ,Host (biology) ,digestive, oral, and skin physiology ,Intercalation (chemistry) ,General Medicine ,Metal ,Transition metal ,visual_art ,Physics::Space Physics ,Polymer chemistry ,Physics::Atomic and Molecular Clusters ,visual_art.visual_art_medium ,Astrophysics::Earth and Planetary Astrophysics - Abstract
The first examples of post-transition metal intercalation into a fullerene host are mercury-loaded derivatives of Na2C60.
- Published
- 2010
- Full Text
- View/download PDF
49. ChemInform Abstract: New Metal Fullerides
- Author
-
Paul F. Henry, A. J. Fowkes, K. M. Allen, A. C. Duggan, Judith M. Fox, and Matthew J. Rosseinsky
- Subjects
Metal ,Chemistry ,visual_art ,Interstitial defect ,visual_art.visual_art_medium ,Charge (physics) ,Nanotechnology ,General Medicine - Abstract
The article is a summary of recent work on the synthesis of new types of metal fulleride designed to examine the influence of various chemical factors on the physical properties of this class of material. Fullende charge spacing, orientations are examined, together with the insertion of less electropositive cations on the interstitial sites.
- Published
- 2010
- Full Text
- View/download PDF
50. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons topoisomerase II
- Author
-
Wenjin Yang, Jo Ann W. Byl, Neil Osheroff, Andrew Conroy, Rachael E. Hawtin, Judith A. Fox, David E. Stockett, Robert S. McDowell, Nguyen Tan, and Michelle R. Arkin
- Subjects
Non-Clinical Medicine ,lcsh:Medicine ,Apoptosis ,Pharmacology ,Quinolones ,Vosaroxin ,Biochemistry ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Delivery Systems ,Oncology/Hematological Malignancies ,lcsh:Science ,Etoposide ,0303 health sciences ,Multidisciplinary ,biology ,Hematology/Acute Myeloid Leukemia ,Quinolone ,Intercalating Agents ,3. Good health ,Chemical Biology/Small Molecule Chemistry ,Oncology ,030220 oncology & carcinogenesis ,Oncology/Breast Cancer ,DNA fragmentation ,Biochemistry/Drug Discovery ,medicine.drug ,Research Article ,G2 Phase ,DNA damage ,medicine.drug_class ,Oncology/Oncology Agents ,Antineoplastic Agents ,DNA Fragmentation ,03 medical and health sciences ,Epipodophyllotoxin ,Cell Line, Tumor ,medicine ,Humans ,Doxorubicin ,Naphthyridines ,Molecular Biology ,030304 developmental biology ,Molecular Biology/DNA Repair ,Topoisomerase ,lcsh:R ,Biochemistry/Chemical Biology of the Cell ,DNA ,Cell Biology ,Thiazoles ,DNA Topoisomerases, Type II ,chemistry ,biology.protein ,lcsh:Q ,DNA Damage ,Pharmacology/Drug Development - Abstract
Background Topoisomerase II is critical for DNA replication, transcription and chromosome segregation and is a well validated target of anti-neoplastic drugs including the anthracyclines and epipodophyllotoxins. However, these drugs are limited by common tumor resistance mechanisms and side-effect profiles. Novel topoisomerase II-targeting agents may benefit patients who prove resistant to currently available topoisomerase II-targeting drugs or encounter unacceptable toxicities. Voreloxin is an anticancer quinolone derivative, a chemical scaffold not used previously for cancer treatment. Voreloxin is completing Phase 2 clinical trials in acute myeloid leukemia and platinum-resistant ovarian cancer. This study defined voreloxin's anticancer mechanism of action as a critical component of rational clinical development informed by translational research. Methods/Principal Findings Biochemical and cell-based studies established that voreloxin intercalates DNA and poisons topoisomerase II, causing DNA double-strand breaks, G2 arrest, and apoptosis. Voreloxin is differentiated both structurally and mechanistically from other topoisomerase II poisons currently in use as chemotherapeutics. In cell-based studies, voreloxin poisoned topoisomerase II and caused dose-dependent, site-selective DNA fragmentation analogous to that of quinolone antibacterials in prokaryotes; in contrast etoposide, the nonintercalating epipodophyllotoxin topoisomerase II poison, caused extensive DNA fragmentation. Etoposide's activity was highly dependent on topoisomerase II while voreloxin and the intercalating anthracycline topoisomerase II poison, doxorubicin, had comparable dependence on this enzyme for inducing G2 arrest. Mechanistic interrogation with voreloxin analogs revealed that intercalation is required for voreloxin's activity; a nonintercalating analog did not inhibit proliferation or induce G2 arrest, while an analog with enhanced intercalation was 9.5-fold more potent. Conclusions/Significance As a first-in-class anticancer quinolone derivative, voreloxin is a toposiomerase II-targeting agent with a unique mechanistic signature. A detailed understanding of voreloxin's molecular mechanism, in combination with its evolving clinical profile, may advance our understanding of structure-activity relationships to develop safer and more effective topoisomerase II-targeted therapies for the treatment of cancer.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.