Network


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

Hotspot


Dive into the research topics where Stephen K. Tahir is active.

Publication


Featured researches published by Stephen K. Tahir.


Nature | 2005

An inhibitor of Bcl-2 family proteins induces regression of solid tumours.

Tilman Oltersdorf; Steven W. Elmore; Alexander R. Shoemaker; Robert C. Armstrong; David J. Augeri; Barbara A. Belli; Milan Bruncko; Thomas L. Deckwerth; Jurgen Dinges; Philip J. Hajduk; Mary K. Joseph; Shinichi Kitada; Stanley J. Korsmeyer; Aaron R. Kunzer; Anthony Letai; Chi Li; Michael J. Mitten; David G. Nettesheim; Shi-Chung Ng; Paul Nimmer; Jacqueline M. O'Connor; Anatol Oleksijew; Andrew M. Petros; John C. Reed; Wang Shen; Stephen K. Tahir; Craig B. Thompson; Kevin J. Tomaselli; Baole Wang; Michael D. Wendt

Proteins in the Bcl-2 family are central regulators of programmed cell death, and members that inhibit apoptosis, such as Bcl-XL and Bcl-2, are overexpressed in many cancers and contribute to tumour initiation, progression and resistance to therapy. Bcl-XL expression correlates with chemo-resistance of tumour cell lines, and reductions in Bcl-2 increase sensitivity to anticancer drugs and enhance in vivo survival. The development of inhibitors of these proteins as potential anti-cancer therapeutics has been previously explored, but obtaining potent small-molecule inhibitors has proved difficult owing to the necessity of targeting a protein–protein interaction. Here, using nuclear magnetic resonance (NMR)-based screening, parallel synthesis and structure-based design, we have discovered ABT-737, a small-molecule inhibitor of the anti-apoptotic proteins Bcl-2, Bcl-XL and Bcl-w, with an affinity two to three orders of magnitude more potent than previously reported compounds. Mechanistic studies reveal that ABT-737 does not directly initiate the apoptotic process, but enhances the effects of death signals, displaying synergistic cytotoxicity with chemotherapeutics and radiation. ABT-737 exhibits single-agent-mechanism-based killing of cells from lymphoma and small-cell lung carcinoma lines, as well as primary patient-derived cells, and in animal models, ABT-737 improves survival, causes regression of established tumours, and produces cures in a high percentage of the mice.


Nature Medicine | 2013

ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets

Andrew J. Souers; Joel D. Leverson; Erwin R. Boghaert; Scott L. Ackler; Nathaniel D. Catron; Jun Chen; Brian D Dayton; H. Ding; Sari H. Enschede; Wayne J. Fairbrother; David C. S. Huang; Sarah G. Hymowitz; Sha Jin; Seong Lin Khaw; Peter Kovar; Lloyd T. Lam; Jackie Lee; Heather Maecker; Kennan Marsh; Kylie D. Mason; Michael J. Mitten; Paul Nimmer; Anatol Oleksijew; Chang H. Park; Cheol-Min Park; Darren C. Phillips; Andrew W. Roberts; Deepak Sampath; John F. Seymour; Morey L. Smith

Proteins in the B cell CLL/lymphoma 2 (BCL-2) family are key regulators of the apoptotic process. This family comprises proapoptotic and prosurvival proteins, and shifting the balance toward the latter is an established mechanism whereby cancer cells evade apoptosis. The therapeutic potential of directly inhibiting prosurvival proteins was unveiled with the development of navitoclax, a selective inhibitor of both BCL-2 and BCL-2–like 1 (BCL-XL), which has shown clinical efficacy in some BCL-2–dependent hematological cancers. However, concomitant on-target thrombocytopenia caused by BCL-XL inhibition limits the efficacy achievable with this agent. Here we report the re-engineering of navitoclax to create a highly potent, orally bioavailable and BCL-2–selective inhibitor, ABT-199. This compound inhibits the growth of BCL-2–dependent tumors in vivo and spares human platelets. A single dose of ABT-199 in three patients with refractory chronic lymphocytic leukemia resulted in tumor lysis within 24 h. These data indicate that selective pharmacological inhibition of BCL-2 shows promise for the treatment of BCL-2–dependent hematological cancers.


Cancer Research | 2008

ABT-263: A Potent and Orally Bioavailable Bcl-2 Family Inhibitor

Christin Tse; Alexander R. Shoemaker; Jessica Adickes; Mark G. Anderson; Jun Chen; Sha Jin; Eric F. Johnson; Kennan Marsh; Michael J. Mitten; Paul Nimmer; Lisa R. Roberts; Stephen K. Tahir; Yu Xiao; Xiufen Yang; Haichao Zhang; Stephen W. Fesik; Saul H. Rosenberg; Steven W. Elmore

Overexpression of the prosurvival Bcl-2 family members (Bcl-2, Bcl-xL, and Mcl-1) is commonly associated with tumor maintenance, progression, and chemoresistance. We previously reported the discovery of ABT-737, a potent, small-molecule Bcl-2 family protein inhibitor. A major limitation of ABT-737 is that it is not orally bioavailable, which would limit chronic single agent therapy and flexibility to dose in combination regimens. Here we report the biological properties of ABT-263, a potent, orally bioavailable Bad-like BH3 mimetic (K(i)s of <1 nmol/L for Bcl-2, Bcl-xL, and Bcl-w). The oral bioavailability of ABT-263 in preclinical animal models is 20% to 50%, depending on formulation. ABT-263 disrupts Bcl-2/Bcl-xL interactions with pro-death proteins (e.g., Bim), leading to the initiation of apoptosis within 2 hours posttreatment. In human tumor cells, ABT-263 induces Bax translocation, cytochrome c release, and subsequent apoptosis. Oral administration of ABT-263 alone induces complete tumor regressions in xenograft models of small-cell lung cancer and acute lymphoblastic leukemia. In xenograft models of aggressive B-cell lymphoma and multiple myeloma where ABT-263 exhibits modest or no single agent activity, it significantly enhances the efficacy of clinically relevant therapeutic regimens. These data provide the rationale for clinical trials evaluating ABT-263 in small-cell lung cancer and B-cell malignancies. The oral efficacy of ABT-263 should provide dosing flexibility to maximize clinical utility both as a single agent and in combination regimens.


Cancer Research | 2007

Influence of Bcl-2 Family Members on the Cellular Response of Small-Cell Lung Cancer Cell Lines to ABT-737

Stephen K. Tahir; Xiufen Yang; Mark G. Anderson; Susan E. Morgan-Lappe; Aparna V. Sarthy; Jun Chen; Robert Warner; Shi-Chung Ng; Stephen W. Fesik; Steve W. Elmore; Saul H. Rosenberg; Christin Tse

ABT-737 is a novel and potent Bcl-2 antagonist with single-agent activity against small-cell lung cancer (SCLC) cell lines. Here, we evaluated the contribution of Bcl-2 family members to the in vitro cellular response of several SCLC cell lines to ABT-737. Relatively higher levels of Bcl-2, Bcl-X(L), Bim and Noxa, and lower levels of Mcl-1 characterized naïve SCLC cell lines that were sensitive to ABT-737. Conversely, a progressive decrease in the relative levels of Bcl-2 and Noxa and a progressive increase in Mcl-1 levels characterized the increased resistance of H146 cells following chronic exposure to ABT-737. Knockdown of Mcl-1 with small interfering RNA sensitized two resistant SCLC cell lines H196 and DMS114 to ABT-737 by enhancing the induction of apoptosis. Likewise, up-regulation of Noxa sensitized H196 cells to ABT-737. Combination treatment with DNA-damaging agents was extremely synergistic with ABT-737 and was associated with the down-regulation of Mcl-1 and the up-regulation of Noxa, Puma, and Bim in H196 cells. Thus, SCLC cells sensitive to ABT-737 expressed the target proteins Bcl-2 and Bcl-X(L), whereas Mcl-1 and factors regulating Mcl-1 function seem to contribute to the overall resistance of SCLC cells to ABT-737. Overall, these observations provide further insight as to the mechanistic bases for ABT-737 efficacy in SCLC and will be helpful for profiling patients and aiding in the rational design of combination therapies.


Journal of Medicinal Chemistry | 2008

Discovery of an orally bioavailable small molecule inhibitor of prosurvival B-cell lymphoma 2 proteins.

Cheol-Min Park; Milan Bruncko; Jessica Adickes; Joy Bauch; Hong Ding; Aaron R. Kunzer; Kennan Marsh; Paul Nimmer; Alexander R. Shoemaker; Xiaohong Song; Stephen K. Tahir; Christin Tse; Xilu Wang; Michael D. Wendt; Xiufen Yang; Haichao Zhang; Stephen W. Fesik; Saul H. Rosenberg; Steven W. Elmore

Overexpression of prosurvival proteins such as Bcl-2 and Bcl-X L has been correlated with tumorigenesis and resistance to chemotherapy, and thus, the development of antagonists of these proteins may provide a novel means for the treatment of cancer. We recently described the discovery of 1 (ABT-737), which binds Bcl-2, Bcl-X L, and Bcl-w with high affinity, shows robust antitumor activity in murine tumor xenograft models, but is not orally bioavailable. Herein, we report that targeted modifications at three key positions of 1 resulted in a 20-fold improvement in the pharmacokinetic/pharmacodynamic relationship (PK/PD) between oral exposure (AUC) and in vitro efficacy in human tumor cell lines (EC 50). The resulting compound, 2 (ABT-263), is orally efficacious in an established xenograft model of human small cell lung cancer, inducing complete tumor regressions in all animals. Compound 2 is currently in multiple phase 1 clinical trials in patients with small cell lung cancer and hematological malignancies.


Science Translational Medicine | 2015

Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy

Joel D. Leverson; Darren C. Phillips; Michael J. Mitten; Erwin R. Boghaert; Stephen K. Tahir; Lisa D. Belmont; Paul Nimmer; Yu Xiao; Xiaoju Max Ma; Kym N. Lowes; Peter Kovar; Jun Chen; Sha Jin; Morey L. Smith; John Xue; Haichao Zhang; Anatol Oleksijew; Terrance J. Magoc; Kedar S. Vaidya; Daniel H. Albert; Jacqueline M. Tarrant; Nghi La; Le Wang; Zhi-Fu Tao; Michael D. Wendt; Deepak Sampath; Saul H. Rosenberg; Chris Tse; David C. S. Huang; Wayne J. Fairbrother

Selective inhibition of BCL-XL synergizes with docetaxel to inhibit the growth of solid tumors but does not inhibit granulopoiesis. A more refined antitumor strategy The BCL-2 family is a group of related proteins that regulate apoptosis in a variety of ways. The success of anticancer treatments often hinges on the ability to induce cancer cell death by apoptosis. As a result, there has been a great deal of interest in developing drugs that can inhibit the antiapoptotic members of the BCL-2 pathway. Unfortunately, some of these drugs are also associated with dose-limiting hematologic toxicities, such as neutropenia. Now, Leverson et al. have used a toolkit of BCL-2 family inhibitors with different specificities to show that specifically inhibiting BCL-XL (one member of this protein family) is effective for killing tumors, but without the common side effects seen with less selective drugs. The BCL-2/BCL-XL/BCL-W inhibitor ABT-263 (navitoclax) has shown promising clinical activity in lymphoid malignancies such as chronic lymphocytic leukemia. However, its efficacy in these settings is limited by thrombocytopenia caused by BCL-XL inhibition. This prompted the generation of the BCL-2–selective inhibitor venetoclax (ABT-199/GDC-0199), which demonstrates robust activity in these cancers but spares platelets. Navitoclax has also been shown to enhance the efficacy of docetaxel in preclinical models of solid tumors, but clinical use of this combination has been limited by neutropenia. We used venetoclax and the BCL-XL–selective inhibitors A-1155463 and A-1331852 to assess the relative contributions of inhibiting BCL-2 or BCL-XL to the efficacy and toxicity of the navitoclax-docetaxel combination. Selective BCL-2 inhibition suppressed granulopoiesis in vitro and in vivo, potentially accounting for the exacerbated neutropenia observed when navitoclax was combined with docetaxel clinically. By contrast, selectively inhibiting BCL-XL did not suppress granulopoiesis but was highly efficacious in combination with docetaxel when tested against a range of solid tumors. Therefore, BCL-XL–selective inhibitors have the potential to enhance the efficacy of docetaxel in solid tumors and avoid the exacerbation of neutropenia observed with navitoclax. These studies demonstrate the translational utility of this toolkit of selective BCL-2 family inhibitors and highlight their potential as improved cancer therapeutics.


Cancer Research | 2006

A Small-Molecule Inhibitor of Bcl-XL Potentiates the Activity of Cytotoxic Drugs In vitro and In vivo

Alex R. Shoemaker; Anatol Oleksijew; Joy Bauch; Barbara A. Belli; Tony Borre; Milan Bruncko; Thomas Deckwirth; David J. Frost; Ken Jarvis; Mary K. Joseph; Kennan Marsh; William J. McClellan; Hugh N. Nellans; Shi-Chung Ng; Paul Nimmer; Jacqueline M. O'Connor; Tilman Oltersdorf; Weiguo Qing; Wang Shen; Jason Stavropoulos; Stephen K. Tahir; Baole Wang; Robert Warner; Haichao Zhang; Stephen W. Fesik; Saul H. Rosenberg; Steven W. Elmore

Inhibition of the prosurvival members of the Bcl-2 family of proteins represents an attractive strategy for the treatment of cancer. We have previously reported the activity of ABT-737, a potent inhibitor of Bcl-2, Bcl-X(L), and Bcl-w, which exhibits monotherapy efficacy in xenograft models of small-cell lung cancer and lymphoma and potentiates the activity of numerous cytotoxic agents. Here we describe the biological activity of A-385358, a small molecule with relative selectivity for binding to Bcl-X(L) versus Bcl-2 (K(i)s of 0.80 and 67 nmol/L for Bcl-X(L) and Bcl-2, respectively). This compound efficiently enters cells and co-localizes with the mitochondrial membrane. Although A-385358 shows relatively modest single-agent cytotoxic activity against most tumor cell lines, it has an EC(50) of <500 nmol/L in cells dependent on Bcl-X(L) for survival. In addition, A-385358 enhances the in vitro cytotoxic activity of numerous chemotherapeutic agents (paclitaxel, etoposide, cisplatin, and doxorubicin) in several tumor cell lines. In A549 non-small-cell lung cancer cells, A-385358 potentiates the activity of paclitaxel by as much as 25-fold. Importantly, A-385358 also potentiated the activity of paclitaxel in vivo. Significant inhibition of tumor growth was observed when A-385358 was added to maximally tolerated or half maximally tolerated doses of paclitaxel in the A549 xenograft model. In tumors, the combination therapy also resulted in a significant increase in mitotic arrest followed by apoptosis relative to paclitaxel monotherapy.


Chemical Biology & Drug Design | 2007

Discovery and Design of Novel HSP90 Inhibitors Using Multiple Fragment-based Design Strategies

Jeffrey R. Huth; Chang Park; Andrew M. Petros; Aaron R. Kunzer; Michael D. Wendt; Xilu Wang; Christopher L. Lynch; Jamey Mack; Kerry M. Swift; Russell A. Judge; Jun Chen; Paul L. Richardson; Sha Jin; Stephen K. Tahir; Edward D. Matayoshi; Sarah A. Dorwin; Uri S. Ladror; Jean M. Severin; Karl A. Walter; Diane Bartley; Stephen W. Fesik; Steven W. Elmore; Philip J. Hajduk

The molecular chaperone HSP90 has been shown to facilitate cancer cell survival by stabilizing key proteins responsible for a malignant phenotype. We report here the results of parallel fragment‐based drug design approaches in the design of novel HSP90 inhibitors. Initial aminopyrimidine leads were elaborated using high‐throughput organic synthesis to yield nanomolar inhibitors of the enzyme. Second site leads were also identified which bound to HSP90 in two distinct conformations, an ‘open’ and ‘closed’ form. Intriguingly, linked fragment approaches targeting both of these conformations were successful in producing novel, micromolar inhibitors. Overall, this study shows that, with only a few fragment hits, multiple lead series can be generated for HSP90 due to the inherent flexibility of the active site. Thus, ample opportunities exist to use these lead series in the development of clinically useful HSP90 inhibitors for the treatment of cancers.


Cancer Chemotherapy and Pharmacology | 2010

The Bcl-2 inhibitor ABT-263 enhances the response of multiple chemotherapeutic regimens in hematologic tumors in vivo

Scott L. Ackler; Michael J. Mitten; Kelly Foster; Anatol Oleksijew; Marion Refici; Stephen K. Tahir; Yu Xiao; Christin Tse; David J. Frost; Stephen W. Fesik; Saul H. Rosenberg; Steven W. Elmore; Alexander R. Shoemaker

PurposeThis study was designed to test the ability of the Bcl-2 family inhibitor ABT-263 to potentiate commonly used chemotherapeutic agents and regimens in hematologic tumor models.MethodsModels of B-cell lymphoma and multiple myeloma were tested in vitro and in vivo with ABT-263 in combination with standard chemotherapeutic regimens, including VAP, CHOP and R-CHOP, as well as single cytotoxic agents including etoposide, rituximab, bortezomib and cyclophosphamide. Alterations in Bcl-2 family member expression patterns were analyzed to define mechanisms of potentiation.ResultsABT-263 was additive with etoposide, vincristine and VAP in vitro in the diffuse large B-cell lymphoma line (DLBCL) DoHH-2, while rituximab potentiated its activity in SuDHL-4. Bortezomib strongly synergized with ABT-263 in the mantle cell lymphoma line Granta 519. Treatment of DoHH-2 with etoposide was associated with an increase in Puma expression, while bortezomib upregulated Noxa expression in Granta 519. Combination of ABT-263 with cytotoxic agents demonstrated superior tumor growth inhibition and delay in multiple models versus cytotoxic therapy alone, along with significant improvements in tumor response rates.ConclusionsInhibition of the Bcl-2 family of proteins by ABT-263 enhances the cytotoxicity of multiple chemotherapeutics in hematologic tumors and represents a promising addition to the therapeutic arsenal for treatment of these diseases.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis and Biological Evaluation of 2-Indolyloxazolines as a New Class of Tubulin Polymerization Inhibitors. Discovery of A-289099 as an Orally Active Antitumor Agent

Qun Li; Keith W. Woods; Akiyo Claiborne; Stephen L. Gwaltney; Kenneth J. Barr; Gang Liu; Laura Gehrke; R. Bruce Credo; Yu Hua Hui; Jang Lee; Robert Warner; Peter Kovar; Michael A. Nukkala; Nicolette A. Zielinski; Stephen K. Tahir; Michael C. Fitzgerald; Ki Kim; Kennan Marsh; David J. Frost; Shi Chung Ng; Saul H. Rosenberg; Hing L. Sham

A series of indole containing oxazolines has been discovered as a result of structural modifications of the lead compound A-105972. The compounds exert their anticancer activity through inhibition of tubulin polymerization by binding at the colchicine site. A-289099 was identified as an orally active antimitotic agent active against various cancer cell lines including those that express the MDR phenotype. The anticancer activity, pharmacokinetics, and an efficient and enantioselective synthesis of A-289099 are described.

Collaboration


Dive into the Stephen K. Tahir's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel D. Leverson

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Hing L. Sham

Thermo Fisher Scientific

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lloyd T. Lam

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Xue

Rockefeller University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge