Network


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

Hotspot


Dive into the research topics where Bonnee Rubinfeld is active.

Publication


Featured researches published by Bonnee Rubinfeld.


PLOS ONE | 2010

Wnt Isoform-Specific Interactions with Coreceptor Specify Inhibition or Potentiation of Signaling by LRP6 Antibodies

Yan Gong; Eric Bourhis; Cecilia Chiu; Scott Stawicki; Venita I. Dealmeida; Bob Y. Liu; Khanhky Phamluong; Tim C. Cao; Richard A. D. Carano; James A. Ernst; Mark Solloway; Bonnee Rubinfeld; Rami N. Hannoush; Yan Wu; Paul Polakis; Mike Costa

β-catenin-dependent Wnt signaling is initiated as Wnt binds to both the receptor FZD and coreceptor LRP5/6, which then assembles a multimeric complex at the cytoplasmic membrane face to recruit and inactivate the kinase GSK3. The large number and sequence diversity of Wnt isoforms suggest the possibility of domain-specific ligand-coreceptor interactions, and distinct binding sites on LRP6 for Wnt3a and Wnt9b have recently been identified in vitro. Whether mechanistically different interactions between Wnts and coreceptors might mediate signaling remains to be determined. It is also not clear whether coreceptor homodimerization induced extracellularly can activate Wnt signaling, as is the case for receptor tyrosine kinases. We generated monoclonal antibodies against LRP6 with the unexpected ability to inhibit signaling by some Wnt isoforms and potentiate signaling by other isoforms. In cell culture, two antibodies characterized further show reciprocal activities on most Wnts, with one antibody antagonizing and the other potentiating. We demonstrate that these antibodies bind to different regions of LRP6 protein, and inhibition of signaling results from blocking Wnt binding. Antibody-mediated dimerization of LRP6 can potentiate signaling only when a Wnt isoform is also able to bind the complex, presumably recruiting FZD. Endogenous autocrine Wnt signaling in different tumor cell lines can be either antagonized or enhanced by the LRP6 antibodies, indicating expression of different Wnt isoforms. As anticipated from the roles of Wnt signaling in cancer and bone development, antibody activities can also be observed in mice for inhibition of tumor growth and in organ culture for enhancement of bone mineral density. Collectively, our results indicate that separate binding sites for different subsets of Wnt isoforms determine the inhibition or potentiation of signaling conferred by LRP6 antibodies. This complexity of coreceptor-ligand interactions may allow for differential regulation of signaling by Wnt isoforms during development, and can be exploited with antibodies to differentially manipulate Wnt signaling in specific tissues or disease states.


Bioconjugate Chemistry | 2011

Impact of drug conjugation on pharmacokinetics and tissue distribution of anti-STEAP1 antibody-drug conjugates in rats.

C. Andrew Boswell; Eduardo E. Mundo; Crystal Zhang; Daniela Bumbaca; Nicole R. Valle; Katherine R. Kozak; Aimee Fourie; Josefa Chuh; Neelima Koppada; Ola Saad; Herman S. Gill; Ben-Quan Shen; Bonnee Rubinfeld; Jay Tibbitts; Surinder Kaur; Frank-Peter Theil; Paul J. Fielder; Leslie A. Khawli; Kedan Lin

Antibody-drug conjugates (ADCs) are designed to combine the exquisite specificity of antibodies to target tumor antigens with the cytotoxic potency of chemotherapeutic drugs. In addition to the general chemical stability of the linker, a thorough understanding of the relationship between ADC composition and biological disposition is necessary to ensure that the therapeutic window is not compromised by altered pharmacokinetics (PK), tissue distribution, and/or potential organ toxicity. The six-transmembrane epithelial antigen of prostate 1 (STEAP1) is being pursued as a tumor antigen target. To assess the role of ADC composition in PK, we evaluated plasma and tissue PK profiles in rats, following a single dose, of a humanized anti-STEAP1 IgG1 antibody, a thio-anti-STEAP1 (ThioMab) variant, and two corresponding thioether-linked monomethylauristatin E (MMAE) drug conjugates modified through interchain disulfide cysteine residues (ADC) and engineered cysteines (TDC), respectively. Plasma PK of total antibody measured by enzyme-linked immunosorbent assay (ELISA) revealed ∼45% faster clearance for the ADC relative to the parent antibody, but no apparent difference in clearance between the TDC and unconjugated parent ThioMab. Total antibody clearances of the two unconjugated antibodies were similar, suggesting minimal effects on PK from cysteine mutation. An ELISA specific for MMAE-conjugated antibody indicated that the ADC cleared more rapidly than the TDC, but total antibody ELISA showed comparable clearance for the two drug conjugates. Furthermore, consistent with relative drug load, the ADC had a greater magnitude of drug deconjugation than the TDC in terms of free plasma MMAE levels. Antibody conjugation had a noticeable, albeit minor, impact on tissue distribution with a general trend toward increased hepatic uptake and reduced levels in other highly vascularized organs. Liver uptakes of ADC and TDC at 5 days postinjection were 2-fold and 1.3-fold higher, respectively, relative to the unmodified antibodies. Taken together, these results indicate that the degree of overall structural modification in anti-STEAP1-MMAE conjugates has a corresponding level of impact on both PK and tissue distribution.


Journal of Biological Chemistry | 2001

Axin-dependent Phosphorylation of the Adenomatous Polyposis Coli Protein Mediated by Casein Kinase 1ε

Bonnee Rubinfeld; David A. Tice; Paul Polakis

Axin and the adenomatous polyposis coli protein (APC) interact to down-regulate the proto-oncogene β-catenin. We show that transposition of an axin-binding site can confer β-catenin regulatory activity to a fragment of APC normally lacking this activity. The fragment containing the axin-binding site also underwent hyperphosphorylation when coexpressed with axin. The phosphorylation did not require glycogen synthase kinase 3β but instead required casein kinase 1ε, which bound directly to axin. Mutation of conserved serine residues in the β-catenin regulatory motifs of APC interfered with both axin-dependent phosphorylation and phosphorylation by CKIε and impaired the ability of APC to regulate β-catenin. These results suggest that the axin-dependent phosphorylation of APC is mediated in part by CKIε and is involved in the regulation of APC function.


Nature Biotechnology | 2006

Identification and immunotherapeutic targeting of antigens induced by chemotherapy

Bonnee Rubinfeld; Archana Upadhyay; Suzanna Clark; Sharon Fong; Victoria Smith; Hartmut Koeppen; Sarajane Ross; Paul Polakis

Cancer cells differ from normal cells in their response to chemotherapy. We exploited this dissimilarity by identifying and targeting tumor-specific, cell-surface proteins whose expression is induced by the chemotherapeutic irinotecan (CPT-11; Camptosar). A cytotoxin-armed antibody reactive with one of these drug-induced surface proteins, the LY6D/E48 antigen, originally identified as the target of a monoclonal antibody reactive with squamous cell carcinomas, caused complete regression of colorectal tumor xenografts in mice treated with CPT-11, whereas either agent alone was less effective. These results suggest that a positive therapeutic index may be generated for other drug combinations by immunotherapeutic targeting of chemotherapy-induced antigens.


Clinical Cancer Research | 2015

Preclinical Development of an Anti-NaPi2b (SLC34A2) Antibody Drug Conjugate as a Therapeutic for Non-Small Cell Lung and Ovarian Cancers

Kedan Lin; Bonnee Rubinfeld; Crystal Zhang; Ron Firestein; Eric Harstad; Leslie Roth; Siao Ping Tsai; Melissa Schutten; Keyang Xu; Maria Hristopoulos; Paul Polakis

Purpose: Antibody–drug conjugates (ADC) selectively deliver a cytotoxic drug to cells expressing an accessible antigenic target. Here, we have appended monomethyl auristatin E (MMAE) to an antibody recognizing the SLC34A2 gene product NaPi2b, the type II sodium–phosphate cotransporter, which is highly expressed on tumor surfaces of the lung, ovary, and thyroid as well as on normal lung pneumocytes. This study evaluated its efficacy and safety in preclinical studies. Experimental Design: The efficacy of anti-NaPi2b ADC was evaluated in mouse ovarian and non–small cell lung cancer (NSCLC) tumor xenograft models, and its toxicity was assessed in rats and cynomolgus monkeys. Results: We show here that an anti-NaPi2b ADC is effective in mouse ovarian and NSCLC tumor xenograft models and well-tolerated in rats and cynomolgus monkeys at levels in excess of therapeutic doses. Despite high levels of expression in normal lung of non-human primate, the cross-reactive ADC exhibited an acceptable safety profile with a dose-limiting toxicity unrelated to normal tissue target expression. The nonproliferative nature of normal pneumocytes, together with the antiproliferative mechanism of MMAE, likely mitigates the potential liability of this normal tissue expression. Conclusions: Overall, our preclinical results suggest that the ADC targeting NaPi2b provides an effective new therapy for the treatment of NSCLC and ovarian cancer and is currently undergoing clinical developments. Clin Cancer Res; 21(22); 5139–50. ©2015 AACR.


PLOS ONE | 2014

Evolutionary Divergence in the Catalytic Activity of the CAM-1, ROR1 and ROR2 Kinase Domains

Travis W. Bainbridge; Venita I. Dealmeida; Anita Izrael-Tomasevic; Cecile Chalouni; Borlan Pan; Joshua Goldsmith; Alia P. Schoen; Gabriel Quinones; Ryan Kelly; Jennie R. Lill; Wendy Sandoval; Mike Costa; Paul Polakis; David Arnott; Bonnee Rubinfeld; James A. Ernst

Receptor tyrosine kinase-like orphan receptors (ROR) 1 and 2 are atypical members of the receptor tyrosine kinase (RTK) family and have been associated with several human diseases. The vertebrate RORs contain an ATP binding domain that deviates from the consensus amino acid sequence, although the impact of this deviation on catalytic activity is not known and the kinase function of these receptors remains controversial. Recently, ROR2 was shown to signal through a Wnt responsive, β-catenin independent pathway and suppress a canonical Wnt/β-catenin signal. In this work we demonstrate that both ROR1 and ROR2 kinase domains are catalytically deficient while CAM-1, the C. elegans homolog of ROR, has an active tyrosine kinase domain, suggesting a divergence in the signaling processes of the ROR family during evolution. In addition, we show that substitution of the non-consensus residues from ROR1 or ROR2 into CAM-1 and MuSK markedly reduce kinase activity, while restoration of the consensus residues in ROR does not restore robust kinase function. We further demonstrate that the membrane-bound extracellular domain alone of either ROR1 or ROR2 is sufficient for suppression of canonical Wnt3a signaling, and that this domain can also enhance Wnt5a suppression of Wnt3a signaling. Based on these data, we conclude that human ROR1 and ROR2 are RTK-like pseudokinases.


Aaps Journal | 2017

Development and Translational Application of an Integrated, Mechanistic Model of Antibody-Drug Conjugate Pharmacokinetics.

Siddharth Sukumaran; Crystal Zhang; Douglas D. Leipold; Ola Saad; Keyang Xu; Kapil Gadkar; Divya Samineni; Bei Wang; Marija Milojic-Blair; Montserrat Carrasco-Triguero; Bonnee Rubinfeld; Paul J. Fielder; Kedan Lin; Saroja Ramanujan

Antibody drug conjugates (ADC), in which small molecule cytotoxic agents are non-specifically linked to antibodies, can enable targeted delivery of chemotherapeutics to tumor cells. ADCs are often produced and administered as a mixture of conjugated antibodies with different drug to antibody ratios (DAR) resulting in complex and heterogeneous disposition kinetics. We developed a mechanism-based platform model that can describe and predict the complex pharmacokinetic (PK) behavior of ADCs with protease-cleavable valine-citrulline (VC) linker linked to Monomethylmonomethyl auristatin F/E by incorporating known mechanisms of ADC disposition. The model includes explicit representation of all DAR species; DAR-dependent sequential deconjugation of the drug, resulting in the conversion of higher DAR to lower DAR species; and DAR-dependent antibody/ADC clearance. PK profiles of multiple analytes (total antibody, drug-conjugated antibody, and/or antibody-conjugated drug) for different ADC molecules and targets in rodents and cynomolgus monkeys were used for model development. The integrated cross-species model was successful in capturing the multi-analyte PK profiles after administration of purified ADCs with defined DAR species and ADCs with mixtures of DAR. Human PK predictions for DSTP3086S (anti-STEAP1-vc-MMAE) with the platform model agreed well with PK (total antibody and antibody-conjugated drug concentrations) measurements in the dose-ranging phase I clinical study. The integrated model is applicable to various other ADCs with different formats, conjugated drugs, and linkers, and provides a valuable tool for the exploration of mechanisms governing disposition of ADCs and enables translational predictions.


Oncotarget | 2016

ImmunoPET helps predicting the efficacy of antibody-drug conjugates targeting TENB2 and STEAP1

Simon-Peter Williams; Annie Ogasawara; Jeff N. Tinianow; Judith E. Flores; David Kan; Jeffrey Lau; Mary Ann Go; Alexander N. Vanderbilt; Herman S. Gill; Li Miao; Joshua Goldsmith; Bonnee Rubinfeld; Weiguang Mao; Ron Firestein; Shang Fan Yu; Jan Marik; Anton G.T.Terwisscha Van Scheltinga

The efficacy of antibody-drug conjugates (ADCs) targeted to solid tumors depends on biological processes that are hard to monitor in vivo. 89Zr-immunoPET of the ADC antibodies could help understand the performance of ADCs in the clinic by confirming the necessary penetration, binding, and internalization. This work studied monomethyl auristatin E (MMAE) ADCs against two targets in metastatic castration-resistant prostate cancer, TENB2 and STEAP1, in four patient-derived tumor models (LuCaP35V, LuCaP70, LuCaP77, LuCaP96.1). Three aspects of ADC biology were measured and compared: efficacy was measured in tumor growth inhibition studies; target expression was measured by immunohistochemistry and flow cytometry; and tumor antibody uptake was measured with 111In-mAbs and gamma counting or with 89Zr-immunoPET. Within each model, the mAb with the highest tumor uptake showed the greatest potency as an ADC. Sensitivity between models varied, with the LuCaP77 model showing weak efficacy despite high target expression and high antibody uptake. Ex vivo analysis confirmed the in vivo results, showing a correlation between expression, uptake and ADC efficacy. We conclude that 89Zr-immunoPET data can demonstrate which ADC candidates achieve the penetration, binding, and internalization necessary for efficacy in tumors sensitive to the toxic payload.


Cancer Research | 2013

Abstract 5619: Preclinical development of anti-SLC34A2 antibody drug conjugate as a therapeutic for non-small cell lung and ovarian cancers.

Kedan Lin; Crystal Zhang; Eric Harstad; Hajime Hiraragi; Willy Solis; Siao Ping Tsai; Keyang Xu; Maria Hristopoulos; Rayna Venook; Bonnee Rubinfeld; Paul Polakis

SLC34A2, also known as type II Na/Pi co-transporters NaPi2b or NaPi3b, is a member of solute carrier protein family SLC34. SLC34A2 is highly expressed on cancers of the lung, ovary and thyroid as well as on normal lung pneumocytes, and is a promising target for antigen-directed therapy. Antibody drug conjugates (ADCs) specify the delivery of a cytotoxic drug to cells expressing an accessible antigenic target. Here we have appended monomethyl auristatin E (MMAE) to an antibody recognizing SLC34A2. Anti-SLC34A2-vc-MMAE inhibits tumor growth in multiple ovarian and non-small cell lung cancer xenograft models that express SLC34A2. Anti-SLC34A2 antibody and its conjugate cross-react with both human and monkey, and toxicity studies in monkeys are conducted to identify both target- and MMAE-related effects. Anti-SLC34A2-vc-MMAE was well tolerated in monkeys at efficacious doses and produced dose-related bone marrow suppression and increased apoptosis and mitotic figures consistent with MMAE-related pharmacology. Despite high levels of expression in normal lung of non-human primate, the cross-reactive ADC exhibited an acceptable safety profile with a dose-limiting toxicity unrelated to normal tissue expression. Overall, our preclinical results suggest that the antibody drug conjugate targeting the SLC34A2 may provide an effective new therapy for the treatment of NSCLC and ovarian cancer. Citation Format: Kedan Lin, Crystal Zhang, Eric Harstad, Hajime Hiraragi, Willy Solis, Siao Ping Tsai, Keyang Xu, Maria Hristopoulos, Rayna Venook, Bonnee Rubinfeld, Paul Polakis. Preclinical development of anti-SLC34A2 antibody drug conjugate as a therapeutic for non-small cell lung and ovarian cancers. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5619. doi:10.1158/1538-7445.AM2013-5619


Molecular Cancer Therapeutics | 2011

Abstract B189: Impact of site of conjugation on efficacy and pharmacokinetics of anti-NaPi3b antibody drug conjugate variants.

Crystal Zhang; Leslie Roth; Ryan Cook; Siao Ping Tsai; Jay Tibbitts; Rayna Venook; Li Miao; Bonnee Rubinfeld; Kedan Lin

Introduction: Optimization of antibody-drug conjugates (ADCs) with therapeutic potential involves several critical parameters, including selection of the antibody, linker, cytotoxic drug, and drug-to-antibody ratio (DAR). To minimize the heterogeneity from conjugation, the antibody can be engineered at different conjugation sites to achieve a defined DAR at specific sites. Anti-NaPi3b-vc-MMAE (anti-NaPi3b-vc-E) is an ADC targeting sodium dependent phosphate transporter expressed in lung and ovarian cancers. Construction and production of the THIOMAB variant of antibody was reported previously. Briefly, a cysteine residue was engineered at Ala118 position of heavy chain (HC) or Val205 position of light chain (LC) to produce its THIOMAB HC and LC variants, respectively. The MMAE conjugates were produced with a drug to antibody ratio (DAR) of two. The purpose of this study was to evaluate the site of conjugation on ADC efficacy and antibody pharmacokinetics. Methods: The anti-tumor activity of anti-Napi3b ADC variants was evaluated in a mouse xenograft model of human ovarian cancer OVCAR3-X2.1 after single dose administration in a range of doses. The unconjugated antibodies including anti-NaPi3b (control) and anti-NaPi3b (HC and LC) and their respective ADC variants were administered intravenously to mice and rats; and the pharmacokinetic (PK) parameters measured by a total antibody assay were estimated by a two-compartmental model. Results: Based on comparisons of tumor growth inhibition, both LC and HC anti-Napi3b-vc-E showed clear dose-dependent inhibitory activity when given at 3 to 12 mg/kg versus the vehicle group. Non-NaPi3b-binding ADCs did not significantly affect tumor growth, indicating targeted anti-tumor activity. The activities of LC and HC variants were similar. Dose proportional pharmacokinetics of total antibody following administration of anti-NaPi3b-vc-E (HC and LC) were observed in mice and rats over the dose range tested. Total antibody clearance of the three unconjugated antibodies (control, thio HC, and thio LC) was comparable. Conjugation with MMAE only modestly increased the total antibody clearance in both LC and HC conjugates when compared to unconjugated antibodies. Overall, PK behavior of the HC and LC variants were similar. Conclusion: The results demonstrated that ADC variants with different sites of conjugation exhibited similar anti-tumor activities, which provided additional choice of conjugation sites for individual antibody. Cysteine mutation in either HC or LC of the unconjugated antibodies did not affect the PK when compared to the control. In addition, conjugation did not significantly change the overall clearance of the antibody, and the site of MMAE conjugations did not have a significant impact on the PK. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr B189.

Collaboration


Dive into the Bonnee Rubinfeld's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge