Network


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

Hotspot


Dive into the research topics where Jean Beebe is active.

Publication


Featured researches published by Jean Beebe.


Clinical Cancer Research | 2005

Combination Therapy Enhances the Inhibition of Tumor Growth with the Fully Human Anti–Type 1 Insulin-Like Growth Factor Receptor Monoclonal Antibody CP-751,871

Bruce D. Cohen; Deborah A. Baker; Catherine Soderstrom; George T. Tkalcevic; Ann Marie Rossi; Penny Miller; Mark W. Tengowski; Faye Wang; Antonio Gualberto; Jean Beebe; James D. Moyer

Purpose: The insulin-like growth factor (IGF) signaling pathway is implicated in cellular mitogenesis, angiogenesis, tumor cell survival, and tumorigenesis. Inhibition of this pathway results in decreased cell growth, inhibition of tumor formation in animal models, and increased apoptosis in cells treated with cytotoxic chemotherapy. We generated and characterized a human monoclonal antibody that targeted the IGF receptor. Experimental Design: By use of XenoMouse technology, we generated CP-751,871, a fully human IgG2 antibody with high affinity (Kd = 1.5 nmol/L) for human IGF-1R and evaluated its biological, pharmacologic, and antitumor properties. Results: This antibody blocks binding of IGF-1 to its receptor (IC50 1.8 nmol/L), IGF-1-induced receptor autophosphorylation (IC50 0.42 nmol/L) and induced the down-regulation of IGF-1R in vitro and in tumor xenografts. The extent of IGF-1R down-regulation in vivo was proportional to CP-751,871 concentrations in the serum of tumor-bearing mice. Pharmacokinetic profiles in cynomolgus monkeys indicated a close to linear increase of exposure following i.v. dosing of antibody in the range of 3 to 100 mg/kg. CP-751,871 showed significant antitumor activity both as a single agent and in combination with Adriamycin, 5-fluorouracil, or tamoxifen in multiple tumor models. A biomarker assay was developed to establish the relationship between circulating antibody concentrations and down-regulation of IGF-1R in peripheral blood cells. The concentration of CP-751,871 required to down-regulate 50% of IGF-1R on peripheral blood cells was 0.3 nmol/L. Conclusion: These data suggest that inhibition of the IGF cascade by use of this monoclonal antibody may be of clinical benefit in the treatment of human cancers.


American Journal of Pathology | 2011

Increased Serum Enzyme Levels Associated with Kupffer Cell Reduction with No Signs of Hepatic or Skeletal Muscle Injury

Zaher A. Radi; Petra Koza-Taylor; Rosonald R. Bell; Leslie Obert; Herbert A. Runnels; Jean Beebe; Michael P. Lawton; Seth Sadis

Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that is responsible for the survival and proliferation of monocytes and the differentiation of monocytes into macrophages, including Kupffer cells (KCs) in the liver. KCs play an important role in the clearance of several serum enzymes, including aspartate aminotransferase and creatine kinase, that are typically elevated as a result of liver or skeletal muscle injury. We used three distinct animal models to investigate the hypothesis that increases in the levels of serum enzymes can be the result of decreases in KCs in the apparent absence of hepatic or skeletal muscle injury. Specifically, neutralizing M-CSF activity via a novel human monoclonal antibody reduced the CD14(+)CD16(+) monocyte population, depleted KCs, and increased aspartate aminotransferase and creatine kinase serum enzyme levels in cynomolgus macaques. In addition, the treatment of rats with clodronate liposomes depleted KCs and led to increased serum enzyme levels, again without evidence of tissue injury. Finally, in the osteopetrotic (Csf1(op)/Csf1(op)) mice lacking functional M-CSF and having reduced levels of KCs, the levels of serum enzymes are higher than in wild-type littermates. Together, these findings support a mechanism for increases in serum enzyme levels through M-CSF regulation of tissue macrophage homeostasis without concomitant histopathological changes in either the hepatic or skeletal system.


Oncogene | 2003

Microarray analysis uncovers retinoid targets in human bronchial epithelial cells

Yan Ma; Petra Koza-Taylor; Debra A. DiMattia; Lynn M. Hames; Haoning Fu; Konstantin H. Dragnev; Tom Turi; Jean Beebe; Sarah J. Freemantle; Ethan Dmitrovsky

Retinoids, the natural and synthetic derivatives of vitamin A, have a role in cancer treatment and prevention. There is a need to reveal mechanisms that account for retinoid response or resistance. This study identified candidate all-trans-retinoic acid (RA) target genes linked to growth suppression in BEAS-2B human bronchial epithelial cells. Microarray analyses were performed using Affymetrix arrays. A total of 11 RA-induced species were validated by reverse transcription polymerase chain reaction (RT-PCR), Western or Northern analyses. Three of these species were novel candidate RA-target genes in human bronchial epithelial cells. These included: placental bone morphogenetic protein (PLAB), polyamine oxidase isoform 1 (PAOh1) and E74-like factor 3 (ELF3). Expression patterns were studied in RA-resistant BEAS-2B-R1 cells. In BEAS-2B-R1 cells, RA dysregulated the expression of the putative lymphocyte G0/G1 switch gene (G0S2), heme oxygenase 1 (HMOX1), tumor necrosis factor-α-induced protein 2 (TNFAIP2), inhibitor of DNA binding 1(Id1), fos-like antigen 1 (FOSL1), transglutaminase 2 (TGM2), asparagine synthetase (ASNS), PLAB, PAOh1 and ELF3, while prominent induction of insulin-like growth-factor-binding protein 6 (IGFBP6) still occurred. In summary, this study identified 11 candidate RA-target genes in human bronchial epithelial cells including three novel species. Expression studies in BEAS-2B-R1 cells indicated that several were directly implicated in RA signaling, since their aberrant expression was linked to RA resistance of human bronchial epithelial cells.


Cancer Research | 2004

Microarray Analyses Uncover UBE1L as a Candidate Target Gene for Lung Cancer Chemoprevention

Ian Pitha-Rowe; W. Jeffrey Petty; Qing Feng; Petra Koza-Taylor; Debra A. DiMattia; Lynn Pinder; Konstantin H. Dragnev; Natalie Memoli; Vincent A. Memoli; Tom Turi; Jean Beebe; Sutisak Kitareewan; Ethan Dmitrovsky

Retinoids, natural and synthetic derivatives of vitamin A, are active in cancer therapy and chemoprevention. We reported previously that all-trans-retinoic acid (RA) treatment prevented carcinogen-induced transformation of immortalized human bronchial epithelial (HBE) cells. To identify cancer chemopreventive mechanisms, immortalized (BEAS-2B), carcinogen-transformed (BEAS-2BNNK), and RA-chemoprevented (BEAS-2BNNK/RA) HBE cells were used to conduct microarray analyses independently. Species increased in chemoprevented as compared with immortalized HBE cells (group I) and those augmented in chemoprevented as compared with transformed HBE cells (group II) included known RA-target genes as well as previously unrecognized RA-target genes in HBE cells. Unexpectedly, both groups were also enriched for interferon-stimulated genes. One interferon-stimulated gene of particular interest was UBE1L, the ubiquitin-activating enzyme E1-like protein. UBE1L expression was also induced after prolonged RA-treatment of immortalized HBE cells. UBE1L mRNA was shown previously as repressed in certain lung cancer cell lines, directly implicating UBE1L in lung carcinogenesis. Notably, UBE1L immunoblot expression was reduced in a subset of malignant as compared with adjacent normal lung tissues that were examined. Immunohistochemical analyses were performed using a new assay developed to detect this species using rabbit polyclonal anti-UBE1L antibodies independently raised against the amino- or carboxyl-termini of UBE1L. Studies done on paraffin-embedded and fixed tissues revealed abundant UBE1L, but low levels of cyclin D1 expression in the normal human bronchial epithelium, indicating an inverse relationship existed between these species. To study this further, cotransfection into HBE cells of wild-type or mutant UBE1L species was accomplished. In a dose-dependent manner, wild-type but not mutant UBE1L species repressed cyclin D1 expression. This implicated UBE1L in a retinoid chemoprevention mechanism involving cyclin D1 repression described previously. Taken together, these findings directly implicate UBE1L as a candidate-pharmacologic target for lung cancer chemoprevention. These findings also provide a mechanistic basis for the tumor suppressive effects of UBE1L through cyclin D1 repression.


The Journal of Clinical Pharmacology | 2014

Anti-IL21 receptor monoclonal antibody (ATR-107): Safety, pharmacokinetics, and pharmacodynamic evaluation in healthy volunteers: a phase I, first-in-human study.

Fei Hua; Gail M. Comer; Lori Stockert; Bo Jin; John Nowak; Susan Pleasic-Williams; David Wunderlich; John B. Cheng; Jean Beebe

Safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of ATR‐107, a fully human monoclonal anti‐IL‐21 receptor (IL‐21R) antibody, administered as ascending single doses, subcutaneously or intravenously, was evaluated in a placebo‐controlled, double‐blind trial in healthy subjects. The dose levels were 3–300 mg by SC and 30–120 mg by IV. The most important adverse events were hypersensitivity reactions occurring in three out of six subjects in 300 mg SC cohort and considered as dose limiting toxicity. More than 75% of the subjects who received ATR‐107 developed anti‐drug antibodies (ADAs), which had no discernible impact on PK or safety. The PK of ATR‐107 appeared to be dose ‐proportional. T1/2 was shorter than typical therapeutic antibodies. Bioavailability of ATR‐107 was about 30%. IL‐21R occupancy was measured in circulating B cells in the 60 and 120 mg IV cohort. The data indicated that single dose of ATR‐107 was able to maximally occupy IL‐21Rs through at least Day 42. Further escalation in the FIH study was halted partially due to the high rates of ADA formation. In conclusion, ATR‐107 had a prolonged PD effect measured by IL‐21R occupancy; was highly immunogenic after single dose administration and had PK properties with rapid clearance and low bioavailability.


Annals of the Rheumatic Diseases | 2017

Efficacy and safety of an interleukin 6 monoclonal antibody for the treatment of systemic lupus erythematosus: a phase II dose-ranging randomised controlled trial

Daniel J. Wallace; Vibeke Strand; Joan T. Merrill; Serghei Popa; Alberto Spindler; Alicia Eimon; Michelle Petri; Josef S Smolen; Joseph Wajdula; Jared Christensen; Cheryl Li; Annette Diehl; Michael Vincent; Jean Beebe; Paul J. Healey; Sudhakar Sridharan

Objectives This phase II trial evaluated the efficacy and safety of an interleukin (IL) 6 monoclonal antibody for systemic lupus erythematosus (SLE). Methods Patients with active disease were randomised to placebo or PF-04236921 10 mg, 50 mg or 200 mg, subcutaneously, every 8 weeks with stable background therapy. SLE Responder Index (SRI-4; primary end point) and British Isles Lupus Assessment Group-based Composite Lupus Assessment (BICLA) were assessed at week 24. Post hoc analysis identified an enriched population based upon planned univariate analyses. Results 183 patients received treatment (placebo, n=45; 10 mg, n=45; 50 mg, n=47; 200 mg, n=46). The 200 mg dose was discontinued due to safety findings and not included in the primary efficacy analysis. The SRI-4 response rates were not significant for any dose compared with placebo; however, the BICLA response rate was significant for 10 mg (p=0.026). The incidence of severe flares was significantly reduced with 10 mg (n=0) and 50 mg (n=2) combined versus placebo (n=8; p<0.01). In patients with greater baseline disease activity (enriched population), the SRI-4 (p=0.004) and BICLA (p=0.012) response rates were significantly different with 10 mg versus placebo. Four deaths (200 mg, n=3; 10 mg, n=1) occurred. The most frequently reported adverse events included headache, nausea and diarrhoea. Conclusions PF-04236921 was not significantly different from placebo for the primary efficacy end point in patients with SLE. Evidence of an effect with 10 mg was seen in a post hoc analysis. Safety was acceptable for doses up to 50 mg as the 200 mg dose was discontinued due to safety findings. Trial registration number NCT01405196; Pre-results.


Advances in Therapy | 2010

PF-03475952: a potent and neutralizing fully human anti-CD44 antibody for therapeutic applications in inflammatory diseases

Herbert A. Runnels; Gregory L. Weber; Jing Min; Elizabeth M. Kudlacz; James F. Zobel; Carol B. Donovan; Mark A. Thiede; Jun Zhang; Robbin Alpert; Michelle A. Salafia; Anthony J. Milici; Douglas Burdette; Rosonald R. Bell; Jean Beebe; Xu Xu

IntroductionCD44 is a cell adhesion molecule believed to play a critical role in T cell and monocyte infiltration in the inflammatory process. The reduction of CD44 expression or its ability to properly interact with its key ligand, hyaluronic acid (HA), inhibits migration and subsequent activation of cells within sites of inflammation. CD44-deficient mice exhibit decreased disease in a mouse arthritis model.MethodsAccordingly, we developed PF-03475952, a fully human IgG2 anti-CD44 monoclonal antibody (mAb).ResultsBinding of PF-03475952 to CD44 inhibits binding of HA and induces loss of CD44 from the cell surface. PF-03475952 also passed a series of safety pharmacology assays designed to assess the risk of the mAb to bind Fc gamma receptors, stimulate cytokine release from human whole blood, and stimulate cytokine release from peripheral blood mononuclear cells (PBMC) using plate-bound antibodies. The latter assay was designed specifically to evaluate the risk of cytokine storm that had been observed with TGN1412 (immunostimulatory CD28 superagonist mAb). PF-003475952 exhibits high-affinity binding to both human and cynomolgus monkey CD44, but does not cross-react with rodent CD44. Thus, a rat anti-mouse CD44 mAb was used to demonstrate a dose-dependent decrease of disease in mouse collagen-induced arthritis. Importantly, efficacy was correlated with >50% loss of cell surface CD44 on circulating cells. Loss of CD44 expression on CD3{su+} lymphocytes was monitored following a single dose of PF-03475952 in cynomolgus monkeys as a pharmacodynamic marker. The recovery of CD44 expression was found to be dose-dependent. PF-03475952 doses of 1, 10, and 100 mg/kg reduced CD44 expression below 50% for 218, 373, and >504 hours, respectively.ConclusionTargeting of CD44 is a unique mechanism of action in the treatment of inflammatory diseases and is expected to reduce joint damage induced by inflammatory mediators, resulting in disease modification in inflammatory diseases such as rheumatoid arthritis.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery and synthesis of novel 4-aminopyrrolopyrimidine Tie-2 kinase inhibitors for the treatment of solid tumors.

Jean Beebe; Martin A. Berliner; Vincent Bernardo; Merin Boehm; Gary Borzillo; Tracey Clark; Bruce D. Cohen; Richard D. Connell; Heather N. Frost; Deborah Gordon; William M. Hungerford; Shefali Kakar; Aaron Kanter; Nandell F. Keene; Elizabeth Knauth; Susan Deborah Lagreca; Yong Lu; Louis Martinez-Alsina; Matthew A. Marx; Joel Morris; Nandini Chaturbhai Patel; Doug Savage; Cathy Soderstrom; Carl Thompson; George T. Tkalcevic; Norma Jacqueline Tom; Felix Vajdos; James J. Valentine; Patrick W. Vincent; Matthew D. Wessel

The synthesis and biological evaluation of novel Tie-2 kinase inhibitors are presented. Based on the pyrrolopyrimidine chemotype, several new series are described, including the benzimidazole series by linking a benzimidazole to the C5-position of the 4-amino-pyrrolopyrimidine core and the ketophenyl series synthesized by incorporating a ketophenyl group to the C5-position. Medicinal chemistry efforts led to potent Tie-2 inhibitors. Compound 15, a ketophenyl pyrrolopyrimidine urea analog with improved physicochemical properties, demonstrated favorable in vitro attributes as well as dose responsive and robust oral tumor growth inhibition in animal models.


Gut | 2017

Randomised trial and open-label extension study of an anti-interleukin-6 antibody in Crohn's disease (ANDANTE I and II)

S. Danese; Severine Vermeire; Paul Hellstern; Remo Panaccione; Gerhard Rogler; Gerald Fraser; Anna Kohn; Pierre Desreumaux; Rupert W. Leong; Gail M. Comer; Fabio Cataldi; Anindita Banerjee; Mary K. Maguire; Cheryl Li; Natalie Rath; Jean Beebe; Stefan Schreiber

Objective Neutralising pro-inflammatory interleukin-6 (IL-6) may effectively treat Crohn’s disease (CD). Effects of PF-04236921, an anti-IL-6 antibody, in adults with CD are reported. Design Parallel-group, dose-ranging, double-blind trial with 4-week screening and 12-week treatment periods. After induction, patients entered 28-week follow-up or 48-week open-label extension (OLE) with 28-week follow-up. Adults with confirmed CD and inadequate response to anti-tumour necrosis factor (TNF) therapy were included. Induction study: 249 patients randomised 1:1:1:1 to placebo, PF-04236921 10, 50 or 200 mg by subcutaneous injection on days 1 and 28. OLE study: PF-04236921 50 mg every 8 weeks up to six doses followed by 28-week follow-up. Results 247 patients were randomised and received treatment in the induction study. The 200 mg dose was discontinued due to safety findings in another study (NCT01405196) and was not included in the primary efficacy analysis. Crohn’s Disease Activity Index (CDAI)-70 response rates with PF-04236921 50 mg were significantly greater than placebo at weeks 8 (49.3% vs 30.6%, P<0.05) and 12 (47.4% vs 28.6%, P<0.05) and met the primary end point. Week 12 CDAI remission rates with PF-04236921 50 mg and placebo were 27.4% and 10.9%, respectively (16.5% difference; P<0.05). 191 subjects received treatment in the OLE. Common treatment-emergent and serious adverse events in both studies included worsening CD, abdominal pain and nasopharyngitis. Conclusions PF-04236921 50 mg induced clinical response and remission in refractory patients with moderate-to-severe CD following failure of anti-TNF therapy. GI abscess and perforation were observed, a specific focus of attention during future clinical development. Trial registration number NCT01287897 and NCT01345318.


Expert Opinion on Therapeutic Patents | 2001

Patent focus on cancer chemotherapeutics. II Angiogenesis agents: April 2000 - September 2000

Richard D. Connell; Jean Beebe

Angiogenesis refers to the formation of capillary blood vessels from existing blood vessels: a process that is believed to be critical for tumour growth and metastasis. Angiogenesis inhibition represents a new approach to cancer chemotherapy with several agents and approaches now entering late clinical development. This review summarises the key aspects of recent patent applications referring to inhibitors of angiogenesis that have been published between April and September 2000. The review covers the main mechanism-based approaches such as MMPI, integrin antagonists, urokinase inhibitors and inhibitors of the growth factor signalling pathways of fibroblast growth factor (FGF), platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and Tie-2/Tek. Applications referring to endogenous inhibitors such as endostatin or angiostatin are also included, as are selected natural products that have data suggesting a link to angiogenesis-specific mechanisms of action.

Collaboration


Dive into the Jean Beebe's collaboration.

Researchain Logo
Decentralizing Knowledge