Network


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

Hotspot


Dive into the research topics where Thomas-Toan Tran is active.

Publication


Featured researches published by Thomas-Toan Tran.


Chemistry & Biology | 2013

Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug Conjugates

Pavel Strop; Shu-Hui Liu; Magdalena Grazyna Dorywalska; Kathy Delaria; Russell Dushin; Thomas-Toan Tran; Wei-Hsien Ho; Santiago E. Farias; Meritxell Galindo Casas; Yasmina Noubia Abdiche; Dahui Zhou; Ramalakshmi Y. Chandrasekaran; Caroline Samain; Carole M. Loo; Andrea Rossi; Mathias Rickert; Stellanie Krimm; Teresa Wong; Sherman Michael Chin; Jessica Yu; Jeanette Dilley; Javier Chaparro-Riggers; Gary Frederick Filzen; Christopher J. O’Donnell; Fang Wang; Jeremy Myers; Jaume Pons; David L. Shelton; Arvind Rajpal

Antibody drug conjugates (ADCs) are a therapeutic class offering promise for cancer therapy. The attachment of cytotoxic drugs to antibodies can result in an effective therapy with better safety potential than nontargeted cytotoxics. To understand the role of conjugation site, we developed an enzymatic method for site-specific antibody drug conjugation using microbial transglutaminase. This allowed us to attach diverse compounds at multiple positions and investigate how the site influences stability, toxicity, and efficacy. We show that the conjugation site has significant impact on ADC stability and pharmacokinetics in a species-dependent manner. These differences can be directly attributed to the position of the linkage rather than the chemical instability, as was observed with a maleimide linkage. With this method, it is possible to produce homogeneous ADCs and tune their properties to maximize the therapeutic window.


Molecular Pharmacology | 2006

Transforming growth factor-β receptor type 1 (TGFβRI) kinase activity but not p38 activation is required for TGFβRI-induced myofibroblast differentiation and profibrotic gene expression

Ann M. Kapoun; Nicholas J. Gaspar; Ying Wang; Debby Damm; Yu-Wang Liu; Gilbert O'Young; Diana Quon; Andrew Lam; Kimberly Munson; Thomas-Toan Tran; Jing Ying Ma; Alison Murphy; Sundeep Dugar; Sarvajit Chakravarty; Andrew A. Protter; Fu-Qiang Wen; Xiangde Liu; Stephen I. Rennard; Linda S. Higgins

Transforming growth factor-β (TGFβ) is a major mediator of normal wound healing and of pathological conditions involving fibrosis, such as idiopathic pulmonary fibrosis. TGFβ also stimulates the differentiation of myofibroblasts, a hallmark of fibrotic diseases. In this study, we examined the underlying processes of TGFβRI kinase activity in myofibroblast conversion of human lung fibroblasts using specific inhibitors of TGFβRI (SD-208) and p38 mitogen-activated kinase (SD-282). We demonstrated that SD-208, but not SD-282, inhibited TGFβ-induced SMAD signaling, myofibroblast transformation, and collagen gel contraction. Furthermore, we extended our findings to a rat bleomycin-induced lung fibrosis model, demonstrating a significant decrease in the number of myofibroblasts at fibroblastic foci in animals treated with SD-208 but not those treated with SD-282. SD-208 also reduced collagen deposition in this in vivo model. Microarray analysis of human lung fibroblasts identified molecular fingerprints of these processes and showed that SD-208 had global effects on reversing TGFβ-induced genes involved in fibrosis, inflammation, cell proliferation, cytoskeletal organization, and apoptosis. These studies also revealed that although the p38 pathway may not be needed for appearance or disappearance of the myofibroblast, it can mediate a subset of inflammatory and fibrogenic events of the myofibroblast during the process of tissue repair and fibrosis. Our findings suggest that inhibitors such as SD-208 may be therapeutically useful in human interstitial lung diseases and pulmonary fibrosis.


Neuro-oncology | 2007

Inhibiting TGF-β signaling restores immune surveillance in the SMA-560 glioma model

Thomas-Toan Tran; Martin Uhl; Jing Ying Ma; Lisa Janssen; Venkataraman Sriram; Steffen Aulwurm; Irene Kerr; Andrew Lam; Heather K. Webb; Ann M. Kapoun; Darin Kizer; Glenn Mcenroe; Barry Hart; Jonathan Axon; Alison Murphy; Sarvajit Chakravarty; Sundeep Dugar; Andrew A. Protter; Linda S. Higgins; Wolfgang Wick; Michael Weller; Darren H. Wong

Transforming growth factor-beta (TGF-beta) is a proinvasive and immunosuppressive cytokine that plays a major role in the malignant phenotype of gliomas. One novel strategy of disabling TGF-beta activity in gliomas is to disrupt the signaling cascade at the level of the TGF-beta receptor I (TGF-betaRI) kinase, thus abrogating TGF-beta-mediated invasiveness and immune suppression. SX-007, an orally active, small-molecule TGF-betaRI kinase inhibitor, was evaluated for its therapeutic potential in cell culture and in an in vivo glioma model. The syngeneic, orthotopic glioma model SMA-560 was used to evaluate the efficacy of SX-007. Cells were implanted into the striatum of VM/Dk mice. Dosing began three days after implantation and continued until the end of the study. Efficacy was established by assessing survival benefit. SX-007 dosed at 20 mg/kg p.o. once daily (q.d.) modulated TGF-beta signaling in the tumor and improved the median survival. Strikingly, approximately 25% of the treated animals were disease-free at the end of the study. Increasing the dose to 40 mg/kg q.d. or 20 mg/kg twice daily did not further improve efficacy. The data suggest that SX-007 can exert a therapeutic effect by reducing TGF-beta-mediated invasion and reversing immune suppression. SX-007 modulates the TGF-beta signaling pathway and is associated with improved survival in this glioma model. Survival benefit is due to reduced tumor invasion and reversal of TGF-beta-mediated immune suppression, allowing for rejection of the tumor. Together, these results suggest that treatment with a TGF-betaRI inhibitor may be useful in the treatment of glioblastoma.


Bioconjugate Chemistry | 2015

Effect of attachment site on stability of cleavable antibody drug conjugates.

Magdalena Grazyna Dorywalska; Pavel Strop; Jody A. Melton-Witt; Adela Hasa-Moreno; Santiago E. Farias; Meritxell Galindo Casas; Kathy Delaria; Victor Lui; Kris Poulsen; Carole M. Loo; Stellanie Krimm; Gary Louis Bolton; Ludivine Moine; Russell Dushin; Thomas-Toan Tran; Shu-Hui Liu; Mathias Rickert; Davide Foletti; David L. Shelton; Jaume Pons; Arvind Rajpal

The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in determining VC-PABC linker stability in mouse plasma, and that the stability of the linker positively correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degradation pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclinical stages of ADC programs. The divergence of linker metabolism in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.


Journal of Cardiovascular Pharmacology | 2004

p38α Mitogen-activated protein kinase inhibition improves cardiac function and reduces myocardial damage in isoproterenol-induced acute myocardial injury in rats

Zhihe Li; Thomas-Toan Tran; Jing Ying Ma; Gilbert O'Young; Ann M. Kapoun; Sarvajit Chakravarty; Sundeep Dugar; George F. Schreiner; Andrew A. Protter

p38 mitogen-activated protein (MAP) kinase is activated during ischemic/hypoxic myocardial injury. However, the role of activated p38 MAP kinase on cardiac function after myocardial injury is not well understood. In the present study, we investigated the cardioprotective effects of p38 MAP kinase inhibition in a rat model of acute myocardial injury, induced by subcutaneous injection of isoproterenol (ISO, 20 mg/kg/d for 3 days). A synthetic p38α MAP kinase inhibitor, SD-282 (40 mg/kg) or vehicle (0.25% Tween 80 in saline) was given intraperitoneally twice a day for 3 days, concomitant with ISO treatment. Cardiac function, systolic blood pressure, gene expression including collagen I and III, fibronectin and COX-2, and the myocardial injury were analyzed. Results showed that administration of SD-282 remarkably improved ISO-induced reduction of cardiac function with increases in ejection fraction (P < 0.001), cardiac output (P < 0.05), stroke volume (P < 0.001), and cardiac index (P < 0.01). SD-282 abolished ISO-induced reduction of systolic blood pressure (106.7 ± 2.2 versus 123.1 ± 5.3 mm Hg, P < 0.05). The ISO-induced expression of COX-2, collagen I and III, and fibronectin genes was reduced significantly (P < 0.05 in all cases) by administration of SD-282. The myocardial injury induced by ISO was significantly reduced by the treatment of SD-282 as judged by the reduction of myocardial necrosis. Data suggest that p38α MAP kinase may be involved in the pathogenesis of cardiac dysfunction in ischemic myocardial injury. Inhibition of this enzyme may improve cardiac function and protect myocardium from ischemic/hypoxic injury that occurs during ischemic heart disease.


Nature Biotechnology | 2015

Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading

Pavel Strop; Kathy Delaria; Davide Foletti; Jody Melton Witt; Adela Hasa-Moreno; Kris Poulsen; Meritxell Galindo Casas; Magdalena Grazyna Dorywalska; Santiago E. Farias; Ariel Pios; Victor Lui; Russell Dushin; Dahui Zhou; Thayalan Navaratnam; Thomas-Toan Tran; Janette Sutton; Kevin Lindquist; Bora Han; Shu-Hui Liu; David L. Shelton; Jaume Pons; Arvind Rajpal

Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading


Molecular Cancer Therapeutics | 2016

Molecular basis of valine-citrulline-PABC linker instability in site-specific ADCs and its mitigation by linker design

Magdalena Grazyna Dorywalska; Russell Dushin; Ludivine Moine; Santiago E. Farias; Dahui Zhou; Thayalan Navaratnam; Victor Lui; Adela Hasa-Moreno; Meritxell Galindo Casas; Thomas-Toan Tran; Kathy Delaria; Shu-Hui Liu; Davide Foletti; Christopher J. O'Donnell; Jaume Pons; David L. Shelton; Arvind Rajpal; Pavel Strop

The degree of stability of antibody–drug linkers in systemic circulation, and the rate of their intracellular processing within target cancer cells are among the key factors determining the efficacy of antibody–drug conjugates (ADC) in vivo. Previous studies demonstrated the susceptibility of cleavable linkers, as well as auristatin-based payloads, to enzymatic cleavage in rodent plasma. Here, we identify Carboxylesterase 1C as the enzyme responsible for the extracellular hydrolysis of valine-citrulline-p-aminocarbamate (VC-PABC)-based linkers in mouse plasma. We further show that the activity of Carboxylesterase 1C towards VC-PABC–based linkers, and consequently the stability of ADCs in mouse plasma, can be effectively modulated by small chemical modifications to the linker. While the introduced modifications can protect the VC-PABC–based linkers from extracellular cleavage, they do not significantly alter the intracellular linker processing by the lysosomal protease Cathepsin B. The distinct substrate preference of the serum Carboxylesterase 1C offers the opportunity to modulate the extracellular stability of cleavable ADCs without diminishing the intracellular payload release required for ADC efficacy. Mol Cancer Ther; 15(5); 958–70. ©2016 AACR.


PLOS ONE | 2015

Site-Dependent Degradation of a Non-Cleavable Auristatin-Based Linker-Payload in Rodent Plasma and Its Effect on ADC Efficacy

Magdalena Grazyna Dorywalska; Pavel Strop; Jody A. Melton-Witt; Adela Hasa-Moreno; Santiago E. Farias; Meritxell Galindo Casas; Kathy Delaria; Victor Lui; Kris Poulsen; Janette Sutton; Gary Louis Bolton; Dahui Zhou; Ludivine Moine; Russell Dushin; Thomas-Toan Tran; Shu-Hui Liu; Mathias Rickert; Davide Foletti; David L. Shelton; Jaume Pons; Arvind Rajpal

The efficacy of an antibody-drug conjugate (ADC) is dependent on the properties of its linker-payload which must remain stable while in systemic circulation but undergo efficient processing upon internalization into target cells. Here, we examine the stability of a non-cleavable Amino-PEG6-based linker bearing the monomethyl auristatin D (MMAD) payload site-specifically conjugated at multiple positions on an antibody. Enzymatic conjugation with transglutaminase allows us to create a stable amide linkage that remains intact across all tested conjugation sites on the antibody, and provides us with an opportunity to examine the stability of the auristatin payload itself. We report a position-dependent degradation of the C terminus of MMAD in rodent plasma that has a detrimental effect on its potency. The MMAD cleavage can be eliminated by either modifying the C terminus of the toxin, or by selection of conjugation site. Both approaches result in improved stability and potency in vitro and in vivo. Furthermore, we show that the MMAD metabolism in mouse plasma is likely mediated by a serine-based hydrolase, appears much less pronounced in rat, and was not detected in cynomolgus monkey or human plasma. Clarifying these species differences and controlling toxin degradation to optimize ADC stability in rodents is essential to make the best ADC selection from preclinical models. The data presented here demonstrate that site selection and toxin susceptibility to mouse plasma degradation are important considerations in the design of non-cleavable ADCs, and further highlight the benefits of site-specific conjugation methods.


Molecular Cancer Therapeutics | 2016

RN927C, a Site-Specific Trop-2 Antibody–Drug Conjugate (ADC) with Enhanced Stability, Is Highly Efficacious in Preclinical Solid Tumor Models

Pavel Strop; Thomas-Toan Tran; Magdalena Grazyna Dorywalska; Kathy Delaria; Russell Dushin; Oi Kwan Wong; Wei-Hsien Ho; Dahui Zhou; Aidong Wu; Eugenia Kraynov; Laura Aschenbrenner; Bora Han; Christopher J. O'Donnell; Jaume Pons; Arvind Rajpal; Dave Shelton; Shu-Hui Liu

Trop-2, also known as TACSTD2, EGP-1, GA733-1, and M1S1, is frequently expressed on a variety of human carcinomas, and its expression is often associated with poor prognosis of the diseases. However, it is also present on the epithelium of several normal tissues. A comprehensively designed Trop-2–targeting antibody–drug conjugate (ADC), balancing both efficacy and toxicity, is therefore necessary to achieve clinical utility. To this end, we developed a cleavable Trop-2 ADC (RN927C) using a site-specific transglutaminase-mediated conjugation method and a proprietary microtubule inhibitor (MTI) linker-payload, PF-06380101. Robust in vitro cytotoxicity of RN927C was observed on a panel of Trop-2–expressing tumor cell lines, with IC50 generally in the subnanomolar range. As expected for an MTI-containing ADC, RN927C readily induced mitotic arrest of treated cells in vitro and in vivo, followed by subsequent cell death. The in vivo efficacy of RN927C was tested in multiple cell line and patient-derived xenograft tumor models, including pancreatic, lung, ovarian, and triple-negative breast tumor types. Single-dose administration of RN927C at 0.75 to 3 mg/kg was generally sufficient to induce sustained regression of Trop-2–expressing tumors and showed superior efficacy over standard treatment with paclitaxel or gemcitabine. Administration of RN927C in nonhuman primate toxicity studies resulted in target-mediated effects in skin and oral mucosa, consistent with Trop-2 expression in these epithelial tissues with minimal, non–dose limiting off-target toxicities. On the basis of the combined efficacy and safety results, RN927C is postulated to have a favorable therapeutic index for treatment of solid tumors. Mol Cancer Ther; 15(11); 2698–708. ©2016 AACR.


Blood Advances | 2017

A novel CXCR4 antagonist IgG1 antibody (PF-06747143) for the treatment of hematologic malignancies

Shu-Hui Liu; Yin Gu; Bernadette Pascual; Zhengming Yan; Max Hallin; Cathy Zhang; Conglin Fan; Wenlian Wang; Justine L. Lam; Mary E. Spilker; Rolla Yafawi; Eileen Blasi; Brett H. Simmons; Nanni Huser; Wei-Hsien Ho; Kevin Lindquist; Thomas-Toan Tran; Jyothirmayee Kudaravalli; Jing-Tyan Ma; Gretchen Jimenez; Ishita Barman; Colleen Brown; Sherman Michael Chin; Maria José Costa; David L. Shelton; Tod Smeal; Valeria R. Fantin; Flavia Pernasetti

The chemokine receptor CXCR4 is highly expressed and associated with poor prognosis in multiple malignancies. Upon engagement by its ligand, CXCL12, CXCR4 triggers intracellular signaling pathways that control trafficking of cells to tissues where the ligand is expressed, such as the bone marrow (BM). In hematologic cancers, CXCR4-driven homing of malignant cells to the BM protective niche is a key mechanism driving disease and therapy resistance. We developed a humanized CXCR4 immunoglobulin G1 (IgG1) antibody (Ab), PF-06747143, which binds to CXCR4 and inhibits CXCL12-mediated signaling pathways, as well as cell migration. In in vivo preclinical studies, PF-06747143 monotherapy rapidly and transiently mobilized cells from the BM into the peripheral blood. In addition, PF-06747143 effectively induced tumor cell death via its Fc constant region-mediated effector function. This Fc-mediated cell killing mechanism not only enhanced antitumor efficacy, but also played a role in reducing the duration of cell mobilization, when compared with an IgG4 version of the Ab, which does not have Fc-effector function. PF-06747143 treatment showed strong antitumor effect in multiple hematologic tumor models including non-Hodgkin lymphoma (NHL), acute myeloid leukemia (AML), and multiple myeloma (MM). Importantly, PF-06747143 synergized with standard-of-care agents in a chemoresistant AML patient-derived xenograft model and in an MM model. These findings suggest that PF-06747143 is a potential best-in-class anti-CXCR4 antagonist for the treatment of hematologic malignancies, including in the resistant setting. PF-06747143 is currently in phase 1 clinical trial evaluation (registered at www.clinicaltrials.gov as #NCT02954653).

Collaboration


Dive into the Thomas-Toan Tran's collaboration.

Top Co-Authors

Avatar

Arvind Rajpal

University of California

View shared research outputs
Top Co-Authors

Avatar

Pavel Strop

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge