Network


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

Hotspot


Dive into the research topics where Reina N. Fuji is active.

Publication


Featured researches published by Reina N. Fuji.


Nature Biotechnology | 2012

Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates

Ben-Quan Shen; Keyang Xu; Luna Liu; Helga Raab; Sunil Bhakta; Margaret Kenrick; Kathryn Parsons-Reponte; Janet Tien; Shang-Fan Yu; Elaine Mai; Dongwei Li; Jay Tibbitts; Jakub Baudys; Ola Saad; Suzie J. Scales; Paul J. Mcdonald; Philip E. Hass; Charles Eigenbrot; Trung Nguyen; Willy Solis; Reina N. Fuji; Kelly Flagella; Darshana Ramesh Patel; Susan D. Spencer; Leslie A. Khawli; Allen Ebens; Wai Lee Wong; Richard Vandlen; Surinder Kaur; Mark X. Sliwkowski

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Clinical Cancer Research | 2010

Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer

Jagath R. Junutula; Kelly Flagella; Richard A. Graham; Kathryn Parsons; Edward Ha; Helga Raab; Sunil Bhakta; Trung Nguyen; Debra L. Dugger; Guangmin Li; Elaine Mai; Gail Lewis Phillips; Hajime Hiraragi; Reina N. Fuji; Jay Tibbitts; Richard Vandlen; Susan D. Spencer; Richard H. Scheller; Paul Polakis; Mark X. Sliwkowski

Purpose: Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examined the in vitro potency as well as in vivo preclinical efficacy and safety profiles of a heterogeneous preparation of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. Experimental Design and Results: To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) molecule was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 molecules per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti–cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equivalent doses (μg/m2) and retained efficacy at equivalent antibody doses (mg/kg). An improved safety profile of >2-fold was observed in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6,000 μg DM1/m2) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6,000 μg DM1/m2). Conclusions: The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clinical development of HER2-positive targeted breast cancer therapies. Clin Cancer Res; 16(19); 4769–78. ©2010 AACR.


Cancer Research | 2009

Antibody-Drug Conjugates for the Treatment of Non–Hodgkin's Lymphoma: Target and Linker-Drug Selection

Andrew Polson; Jill Calemine-Fenaux; Pamela Chan; Wesley Chang; Erin H. Christensen; Suzanna Clark; Frederic J. de Sauvage; Dan L. Eaton; Kristi Elkins; J. Michael Elliott; Gretchen Frantz; Reina N. Fuji; Alane Gray; Kristin Harden; Gladys Ingle; Noelyn M. Kljavin; Hartmut Koeppen; Christopher P. Nelson; Saileta Prabhu; Helga Raab; Sarajane Ross; Jean-Philippe Stephan; Suzie J. Scales; Susan D. Spencer; Richard Vandlen; Bernd Wranik; Shang-Fan Yu; Bing Zheng; Allen Ebens

Antibody-drug conjugates (ADC), potent cytotoxic drugs covalently linked to antibodies via chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Here, we systematically examine the potential targets and linker-drug combinations that could provide an optimal ADC for the treatment for non-Hodgkins lymphoma. We identified seven antigens (CD19, CD20, CD21, CD22, CD72, CD79b, and CD180) for potential treatment of non-Hodgkins lymphoma with ADCs. ADCs with cleavable linkers mediated in vivo efficacy via all these targets; ADCs with uncleavable linkers were only effective when targeted to CD22 and CD79b. In target-independent safety studies in rats, the uncleavable linker ADCs showed reduced toxicity, presumably due to the reduced release of free drug or other toxic metabolites into the circulation. Thus, our data suggest that ADCs with cleavable linkers work on a broad range of targets, and for specific targets, ADCs with uncleavable linkers provide a promising opportunity to improve the therapeutic window for ADCs in humans.


Science Translational Medicine | 2013

Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier.

Jessica Couch; Y. Joy Yu; Yin Zhang; Jacqueline M. Tarrant; Reina N. Fuji; William J. Meilandt; Hilda Solanoy; Raymond K. Tong; Kwame Hoyte; Wilman Luk; Yanmei Lu; Kapil Gadkar; Saileta Prabhu; Benjamin A. Ordonia; Quyen Nguyen; Yuwen Lin; Zhonghua Lin; Mercedesz Balazs; Kimberly Scearce-Levie; James A. Ernst; Mark S. Dennis; Ryan J. Watts

The safety of therapeutic bispecific antibodies that use TfR for delivery to the brain can be improved by reducing affinity for TfR and eliminating antibody effector function. Averting Roadblocks En Route to the Brain The blood-brain barrier represents a formidable blockade preventing therapeutic antibody delivery into the brain. Bispecific antibodies using the transferrin receptor (TfR) have shown promise for boosting therapeutic antibody uptake into the brain. Although TfR can act as a molecular lift to promote brain uptake, little is known about the safety ramifications of this approach. Building on a pair of studies published in Science Translational Medicine, Couch and colleagues now report that when mice were dosed with therapeutic TfR antibodies, the animals showed acute clinical reactions and a reduction in immature red blood cells, known as reticulocytes. TfR bispecific antibodies engineered to lack Fc interactions with immune cells eliminated adverse acute clinical reactions and reduced reticulocyte loss; the extent of reticulocyte loss was also influenced by binding to TfR and interaction with the complement cascade. Because reticulocytes express high levels of TfR, other cell types that express high levels of TfR were also investigated. The authors observed, for example, that the blood-brain barrier remained completely intact after TfR antibodies were administered to mice, despite the high expression of TfR in brain endothelial cells. Finally, multiple doses of TfR/BACE1 bispecific antibodies reduced amyloid-β, a toxic protein implicated in Alzheimer’s disease, with minimal sustained toxicity. Investigation of monkey and human TfR levels in circulating reticulocytes suggested that loss of these cells may be less likely to occur in primates than in mice. The translational implications of these discoveries suggest that the blood-brain barrier is not the only obstacle to surmount on the way to the brain, at least when using TfR as a molecular lift. Bispecific antibodies using the transferrin receptor (TfR) have shown promise for boosting antibody uptake in brain. Nevertheless, there are limited data on the therapeutic properties including safety liabilities that will enable successful development of TfR-based therapeutics. We evaluate TfR/BACE1 bispecific antibody variants in mouse and show that reducing TfR binding affinity improves not only brain uptake but also peripheral exposure and the safety profile of these antibodies. We identify and seek to address liabilities of targeting TfR with antibodies, namely, acute clinical signs and decreased circulating reticulocytes observed after dosing. By eliminating Fc effector function, we ameliorated the acute clinical signs and partially rescued a reduction in reticulocytes. Furthermore, we show that complement mediates a residual decrease in reticulocytes observed after Fc effector function is eliminated. These data raise important safety concerns and potential mitigation strategies for the development of TfR-based therapies that are designed to cross the blood-brain barrier.


Journal of Medicinal Chemistry | 2012

Discovery of highly potent, selective, and brain-penetrable leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors.

Anthony A. Estrada; Xingrong Liu; Charles Baker-Glenn; Alan Beresford; Daniel J. Burdick; Mark Stuart Chambers; Bryan K. Chan; Huifen Chen; Xiao Ding; Antonio G. DiPasquale; Sara L. Dominguez; Jennafer Dotson; Jason Drummond; Michael Flagella; Sean P. Flynn; Reina N. Fuji; Andrew Gill; Janet Gunzner-Toste; Seth F. Harris; Timothy P. Heffron; Tracy Kleinheinz; Donna W. Lee; Claire E. Le Pichon; Joseph P. Lyssikatos; Andrew D. Medhurst; John Moffat; Susmith Mukund; Kevin Nash; Kimberly Scearce-Levie; Zejuan Sheng

There is a high demand for potent, selective, and brain-penetrant small molecule inhibitors of leucine-rich repeat kinase 2 (LRRK2) to test whether inhibition of LRRK2 kinase activity is a potentially viable treatment option for Parkinsons disease patients. Herein we disclose the use of property and structure-based drug design for the optimization of highly ligand efficient aminopyrimidine lead compounds. High throughput in vivo rodent cassette pharmacokinetic studies enabled rapid validation of in vitro-in vivo correlations. Guided by this data, optimal design parameters were established. Effective incorporation of these guidelines into our molecular design process resulted in the discovery of small molecule inhibitors such as GNE-7915 (18) and 19, which possess an ideal balance of LRRK2 cellular potency, broad kinase selectivity, metabolic stability, and brain penetration across multiple species. Advancement of GNE-7915 into rodent and higher species toxicity studies enabled risk assessment for early development.


Leukemia | 2010

Anti-CD22-MCC-DM1: An antibody-drug conjugate with a stable linker for the treatment of non-Hodgkin's lymphoma

Andrew G. Polson; Marna Williams; A. M. Gray; Reina N. Fuji; Kirsten Achilles Poon; Jacqueline McBride; Helga Raab; T. Januario; MaryAnn Go; Jeffrey Lau; Shang-Fan Yu; Changchun Du; Franklin Fuh; C. Tan; Y. Wu; W.-C. Liang; Saileta Prabhu; J.-P. Stephan; J.-A. Hongo; Randall Dere; Rong Deng; M. Cullen; R de Tute; Fiona Bennett; Andy C. Rawstron; Andrew Jack; Allen Ebens

Antibody-drug conjugates (ADCs) are potent cytotoxic drugs linked to antibodies through chemical linkers, and allow specific targeting of drugs to neoplastic cells. The expression of CD22 is limited to B-cells, and we show that CD22 is expressed on the vast majority of non-Hodgkins lymphomas (NHLs). An ideal target for an ADC for the treatment of NHL would have limited expression outside the B-cell compartment and be highly effective against NHL. We generated an ADC consisting of a humanized anti-CD22 antibody conjugated to the anti-mitotic agent maytansine with a stable linker (anti-CD22-MCC-DM1). Anti-CD22-MCC-DM1 was broadly effective in in vitro killing assays on NHL B-cell lines. We did not find a strong correlation between in vitro potency and CD22 surface expression, internalization of ADC or sensitivity to free drug. We show that anti-CD22-MCC-DM1 was capable of inducing complete tumor regression in NHL xenograft mouse models. Further, anti-CD22-MCC-DM1 was well tolerated in cynomolgus monkeys and substantially decreased circulating B-cells as well as follicle size and germinal center formation in lymphoid organs. These results suggest that anti-CD22-MCC-DM1 has an efficacy, safety and pharmacodynamic profile that support its use as a treatment for NHL.


Science Translational Medicine | 2015

Effect of selective LRRK2 kinase inhibition on nonhuman primate lung

Reina N. Fuji; Michael Flagella; Miriam Baca; Marco A. S. Baptista; Jens Brodbeck; Bryan K. Chan; Brian K. Fiske; Lee Honigberg; Adrian M. Jubb; Paula Katavolos; Donna W. Lee; Sock-Cheng Lewin-Koh; Tori Lin; Xingrong Liu; Shannon Liu; Joseph P. Lyssikatos; Jennifer O'Mahony; Mike Reichelt; Merone Roose-Girma; Zejuan Sheng; Todd Sherer; Ashley Smith; Margaret Solon; Zachary Kevin Sweeney; Jacqueline M. Tarrant; Alison Urkowitz; Søren Warming; Murat Yaylaoglu; Shuo Zhang; Haitao Zhu

LRRK2 kinase inhibitors, under development for Parkinson’s disease, have an effect on type II pneumocytes in nonhuman primate lung, suggesting that pulmonary toxicity may be a critical safety liability. A lung phenotype for LRRK2 inhibitors Human genetic evidence implicates leucine-rich repeat kinase 2 (LRRK2) as a high-priority drug target for Parkinson’s disease. However, the benefit and risk of inhibiting the kinase activity of LRRK2 is unknown and is currently untested in humans. Using two selective LRRK2 kinase inhibitors, Fuji et al. report a safety liability in nonhuman primates characterized by morphological changes in lung. These results are consistent with observations in mice lacking LRRK2. These safety observations offer a cautionary note for pharmacological modulation of LRRK2 in humans. Inhibition of the kinase activity of leucine-rich repeat kinase 2 (LRRK2) is under investigation as a possible treatment for Parkinson’s disease. However, there is no clinical validation as yet, and the safety implications of targeting LRRK2 kinase activity are not well understood. We evaluated the potential safety risks by comparing human and mouse LRRK2 mRNA tissue expression, by analyzing a Lrrk2 knockout mouse model, and by testing selective brain-penetrating LRRK2 kinase inhibitors in multiple species. LRRK2 mRNA tissue expression was comparable between species. Phenotypic analysis of Lrrk2 knockout mice revealed morphologic changes in lungs and kidneys, similar to those reported previously. However, in preclinical toxicity assessments in rodents, no pulmonary or renal changes were induced by two distinct LRRK2 kinase inhibitors. Both of these kinase inhibitors induced abnormal cytoplasmic accumulation of secretory lysosome-related organelles known as lamellar bodies in type II pneumocytes of the lung in nonhuman primates, but no lysosomal abnormality was observed in the kidney. The pulmonary change resembled the phenotype of Lrrk2 knockout mice, suggesting that this was LRRK2-mediated rather than a nonspecific or off-target effect. A biomarker of lysosomal dysregulation, di-docosahexaenoyl (22:6) bis(monoacylglycerol) phosphate (di-22:6-BMP), was also decreased in the urine of Lrrk2 knockout mice and nonhuman primates treated with LRRK2 kinase inhibitors. Our results suggest a role for LRRK2 in regulating lysosome-related lamellar bodies and that pulmonary toxicity may be a critical safety liability for LRRK2 kinase inhibitors in patients.


Journal of Medicinal Chemistry | 2014

Discovery of Highly Potent, Selective, and Brain-Penetrant Aminopyrazole Leucine-Rich Repeat Kinase 2 (LRRK2) Small Molecule Inhibitors

Anthony A. Estrada; Bryan K. Chan; Charles Baker-Glenn; Alan Beresford; Daniel J. Burdick; Mark Stuart Chambers; Huifen Chen; Sara L. Dominguez; Jennafer Dotson; Jason Drummond; Michael Flagella; Reina N. Fuji; Andrew Gill; Jason S. Halladay; Seth F. Harris; Timothy P. Heffron; Tracy Kleinheinz; Donna W. Lee; Claire E. Le Pichon; Xingrong Liu; Joseph P. Lyssikatos; Andrew D. Medhurst; John Moffat; Kevin Nash; Kimberly Scearce-Levie; Zejuan Sheng; Daniel Shore; Susan Wong; Shuo Zhang; Xiaolin Zhang

Leucine-rich repeat kinase 2 (LRRK2) has drawn significant interest in the neuroscience research community because it is one of the most compelling targets for a potential disease-modifying Parkinsons disease therapy. Herein, we disclose structurally diverse small molecule inhibitors suitable for assessing the implications of sustained in vivo LRRK2 inhibition. Using previously reported aminopyrazole 2 as a lead molecule, we were able to engineer structural modifications in the solvent-exposed region of the ATP-binding site that significantly improve human hepatocyte stability, rat free brain exposure, and CYP inhibition and induction liabilities. Disciplined application of established optimal CNS design parameters culminated in the rapid identification of GNE-0877 (11) and GNE-9605 (20) as highly potent and selective LRRK2 inhibitors. The demonstrated metabolic stability, brain penetration across multiple species, and selectivity of these inhibitors support their use in preclinical efficacy and safety studies.


British Journal of Pharmacology | 2013

An integrated approach to identify normal tissue expression of targets for antibody‐drug conjugates: case study of TENB2

C. Andrew Boswell; Eduardo E. Mundo; Ron Firestein; Crystal Zhang; Weiguang Mao; Herman S. Gill; Cynthia Young; Nina Ljumanovic; Shannon Stainton; Sheila Ulufatu; Aimee Fourie; Katherine R. Kozak; Reina N. Fuji; Paul Polakis; Leslie A. Khawli; Kedan Lin

The success of antibody‐drug conjugates (ADCs) depends on the therapeutic window rendered by the differential expression between normal and pathological tissues. The ability to identify and visualize target expression in normal tissues could reveal causes for target‐mediated clearance observed in pharmacokinetic characterization. TENB2 is a prostate cancer target associated with the progression of poorly differentiated and androgen‐independent tumour types, and ADCs specific for TENB2 are candidate therapeutics. The objective of this study was to locate antigen expression of TENB2 in normal tissues, thereby elucidating the underlying causes of target‐mediated clearance.


British Journal of Pharmacology | 2012

The successes and limitations of preclinical studies in predicting the pharmacodynamics and safety of cell-surface-targeted biological agents in patients

Andrew G. Polson; Reina N. Fuji

To improve drug development outcomes, it is important to review when preclinical pharmacodynamic and safety models have successfully predicted human responses and when they have not. In a recent issue of the BJP, Bugelski and Martin examined the concordance between preclinical and human data for biopharmaceuticals targeted to cell‐surface proteins. The cases are interesting and several trends emerge. The pharmacodynamics of biopharmaceuticals in non‐human primates is largely predictive; the use of surrogates in rodents may be similarly predictive, allowing for more conservative use of non‐human primates. While overall concordance of preclinical toxicology data and clinical safety was poor, this is largely a reflection of the immunomodulatory biology of the majority of the biopharmaceuticals evaluated. The examples show that adverse effects in animals that were the result of direct and/or exaggerated pharmacology were modelled well, but that specific infections or other indirect outcomes of immunomodulation, along with cytokine‐related events, were not modelled well in preclinical studies.

Collaboration


Dive into the Reina N. Fuji's collaboration.

Researchain Logo
Decentralizing Knowledge