Network


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

Hotspot


Dive into the research topics where Francisco Adrian is active.

Publication


Featured researches published by Francisco Adrian.


Nature | 2010

Targeting Bcr-Abl by combining allosteric with ATP-binding-site inhibitors

Jianming Zhang; Francisco Adrian; Wolfgang Jahnke; Sandra W. Cowan-Jacob; Allen Li; Roxana E. Iacob; Taebo Sim; John T. Powers; Christine Dierks; Fangxian Sun; Gui Rong Guo; Qiang Ding; Barun Okram; Yongmun Choi; Amy Wojciechowski; Xianming Deng; Guoxun Liu; Gabriele Fendrich; André Strauss; Navratna Vajpai; Stephan Grzesiek; Tove Tuntland; Yi Liu; Badry Bursulaya; Mohammad Azam; Paul W. Manley; John R. Engen; George Q. Daley; Markus Warmuth; Nathanael S. Gray

In an effort to find new pharmacological modalities to overcome resistance to ATP-binding-site inhibitors of Bcr–Abl, we recently reported the discovery of GNF-2, a selective allosteric Bcr–Abl inhibitor. Here, using solution NMR, X-ray crystallography, mutagenesis and hydrogen exchange mass spectrometry, we show that GNF-2 binds to the myristate-binding site of Abl, leading to changes in the structural dynamics of the ATP-binding site. GNF-5, an analogue of GNF-2 with improved pharmacokinetic properties, when used in combination with the ATP-competitive inhibitors imatinib or nilotinib, suppressed the emergence of resistance mutations in vitro, displayed additive inhibitory activity in biochemical and cellular assays against T315I mutant human Bcr–Abl and displayed in vivo efficacy against this recalcitrant mutant in a murine bone-marrow transplantation model. These results show that therapeutically relevant inhibition of Bcr–Abl activity can be achieved with inhibitors that bind to the myristate-binding site and that combining allosteric and ATP-competitive inhibitors can overcome resistance to either agent alone.


Proceedings of the National Academy of Sciences of the United States of America | 2008

In silico activity profiling reveals the mechanism of action of antimalarials discovered in a high-throughput screen

David Plouffe; Achim Brinker; Case W. McNamara; Kerstin Henson; Nobutaka Kato; Kelli Kuhen; Advait Nagle; Francisco Adrian; Jason Matzen; Paul Anderson; Tae-gyu Nam; Nathanael S. Gray; Arnab K. Chatterjee; Jeff Janes; S. Frank Yan; Richard Trager; Jeremy S. Caldwell; Peter G. Schultz; Yingyao Zhou; Elizabeth A. Winzeler

The growing resistance to current first-line antimalarial drugs represents a major health challenge. To facilitate the discovery of new antimalarials, we have implemented an efficient and robust high-throughput cell-based screen (1,536-well format) based on proliferation of Plasmodium falciparum (Pf) in erythrocytes. From a screen of ≈1.7 million compounds, we identified a diverse collection of ≈6,000 small molecules comprised of >530 distinct scaffolds, all of which show potent antimalarial activity (<1.25 μM). Most known antimalarials were identified in this screen, thus validating our approach. In addition, we identified many novel chemical scaffolds, which likely act through both known and novel pathways. We further show that in some cases the mechanism of action of these antimalarials can be determined by in silico compound activity profiling. This method uses large datasets from unrelated cellular and biochemical screens and the guilt-by-association principle to predict which cellular pathway and/or protein target is being inhibited by select compounds. In addition, the screening method has the potential to provide the malaria community with many new starting points for the development of biological probes and drugs with novel antiparasitic activities.


Current Opinion in Oncology | 2007

Ba/F3 cells and their use in kinase drug discovery.

Markus Warmuth; Sung Joon Kim; Xiang-ju Gu; Gang Xia; Francisco Adrian

Purpose of review Due to their ability to function as dominant oncogenes, protein kinases have become favored targets in the quest for ‘molecularly-targeted’ cancer chemotherapeutics. The discovery of a large number of cancer-associated mutations in the kinome, and the progress in developing specific small-molecule kinase inhibitors has increased the need for accurate, reproducible, and efficient kinase activity-dependent cellular assay systems. Recent findings Ba/F3, a murine interleukin-3 dependent pro-B cell line is increasingly popular as a model system for assessing both the potency and downstream signaling of kinase oncogenes, and the ability of small-molecule kinase inhibitors to block kinase activity. Facilitated by their growth properties, Ba/F3 cells have recently been adapted to high-throughput assay formats for compound profiling. Further, several published approaches show promise in predicting resistance to small-molecule kinase inhibitors elicited by point mutations interfering with inhibitor binding. Summary Ba/F3 cells are an increasingly popular tool in kinase drug discovery. The ability to test the transforming capacity of newly identified kinase mutations, and to profile drug candidates and compound libraries in high-throughput fashion, combined with the use of Ba/F3 cells to predict clinical resistance will greatly facilitate developments in this field.


Cancer Research | 2010

The ITK-SYK Fusion Oncogene Induces a T-Cell Lymphoproliferative Disease in Mice Mimicking Human Disease

Christine Dierks; Francisco Adrian; Paul Fisch; Hong Ma; Helga Maurer; Dieter Herchenbach; Christine Ulrike Forster; Clara Sprissler; Guoxun Liu; Sabine Rottmann; Gui-Rong Guo; Zirlik Katja; Hendrik Veelken; Markus Warmuth

Peripheral T-cell lymphomas (PTCL) constitute a major treatment problem with high mortality rates due to the minimal effectiveness of conventional chemotherapy. Recent findings identified ITK-SYK as the first recurrent translocation in 17% of unspecified PTCLs and showed the overexpression of SYK in more than 90% of PTCLs. Here, we show that the expression of ITK-SYK in the bone marrow of BALB/c mice causes a T-cell lymphoproliferative disease in all transplanted mice within 8 weeks after transplantation. The disease was characterized by the infiltration of spleen, lymph nodes, bone marrow, and skin with CD3+CD4+CD8- and CD3+CD4-CD8- ITK-SYK-positive T-cells accompanied by a systemic inflammatory reaction with upregulation of interleukin 5 and INF-gamma. ITK-SYK-positive T-cells showed enhanced apoptosis resistance and INF-gamma production in vitro. The disease was serially transplantable, inducing clonal T-cell expansion in secondary recipients. The action of ITK-SYK in vivo was dependent on SYK kinase activity and disease development could be inhibited by the treatment of mice with SYK inhibitors. Interestingly, the translocation of ITK-SYK from the membrane to the cytoplasm, using a point mutation in the pleckstrin homology domain (ITK-SYK R29C), did not abolish, but rather, enhanced disease development in transplanted mice. CBL binding was strongly enhanced in membrane-associated ITK-SYK E42K and was causative for delayed disease development. Our results show that ITK-SYK causes a T-cell lymphoproliferative disease in mice, supporting its role in T-cell lymphoma development in humans. Therefore, pharmacologic inhibition of SYK in patients with U-PTCLs carrying the ITK-SYK fusion protein might be an effective treatment strategy.


ACS Medicinal Chemistry Letters | 2012

Discovery of GNF-5837, a Selective TRK Inhibitor with Efficacy in Rodent Cancer Tumor Models

Pam Albaugh; Yi Fan; Yuan Mi; Fangxian Sun; Francisco Adrian; Nanxin Li; Yong Jia; Yelena Sarkisova; Andreas Kreusch; Tami Hood; Min Lu; Guoxun Liu; Shenlin Huang; Zuosheng Liu; Jon Loren; Tove Tuntland; Donald S. Karanewsky; H. Martin Seidel; Valentina Molteni

Neurotrophins and their receptors (TRKs) play key roles in the development of the nervous system and the maintenance of the neural network. Accumulating evidence points to their role in malignant transformations, chemotaxis, metastasis, and survival signaling and may contribute to the pathogenesis of a variety of tumors of both neural and non-neural origin. By screening the GNF kinase collection, a series of novel oxindole inhibitors of TRKs were identified. Optimization led to the identification of GNF-5837 (22), a potent, selective, and orally bioavailable pan-TRK inhibitor that inhibited tumor growth in a mouse xenograft model derived from RIE cells expressing both TRKA and NGF. The properties of 22 make it a good tool for the elucidation of TRK biology in cancer and other nononcology indications.


Biochimica et Biophysica Acta | 2010

Inhibitors of the Abl kinase directed at either the ATP- or myristate-binding site.

Doriano Fabbro; Paul W. Manley; Wolfgang Jahnke; Janis Liebetanz; Alexandra Szyttenholm; Gabriele Fendrich; André Strauss; Jianming Zhang; Nathanael S. Gray; Francisco Adrian; Markus Warmuth; Xavier Francois Andre Pelle; Robert Martin Grotzfeld; Frederic Berst; Andreas Marzinzik; Sandra W. Cowan-Jacob; Pascal Furet

The ATP-competitive inhibitors dasatinib and nilotinib, which bind to catalytically different conformations of the Abl kinase domain, have recently been approved for the treatment of imatinib-resistant CML. These two new drugs, albeit very efficient against most of the imatinib-resistant mutants of Bcr-Abl, fail to effectively suppress the Bcr-Abl activity of the T315I (or gatekeeper) mutation. Generating new ATP site-binding drugs that target the T315I in Abl has been hampered, amongst others, by target selectivity, which is frequently an issue when developing ATP-competitive inhibitors. Recently, using an unbiased cellular screening approach, GNF-2, a non-ATP-competitive inhibitor, has been identified that demonstrates cellular activity against Bcr-Abl transformed cells. The exquisite selectivity of GNF-2 is due to the finding that it targets the myristate binding site located near the C-terminus of the Abl kinase domain, as demonstrated by genetic approaches, solution NMR and X-ray crystallography. GNF-2, like myristate, is able to induce and/or stabilize the clamped inactive conformation of Abl analogous to the SH2-Y527 interaction of Src. The molecular mechanism for allosteric inhibition by the GNF-2 inhibitor class, and the combined effects with ATP-competitive inhibitors such as nilotinib and imatinib on wild-type Abl and imatinib-resistant mutants, in particular the T315I gatekeeper mutant, are reviewed.


Journal of Medicinal Chemistry | 2010

Expanding the diversity of allosteric bcr-abl inhibitors.

Xianming Deng; Barun Okram; Qiang Ding; Jianming Zhang; Yongmun Choi; Francisco Adrian; Amy Wojciechowski; Guobao Zhang; Jianwei Che; Badry Bursulaya; Sandra W. Cowan-Jacob; Gabriele Rummel; Taebo Sim; Nathanael S. Gray

Inhibition of Bcr-Abl kinase activity by imatinib for the treatment of chronic myeloid leukemia (CML) currently serves as the paradigm for targeting dominant oncogenes with small molecules. We recently reported the discovery of GNF-2 (1) and GNF-5 (2) as selective non-ATP competitive inhibitors of cellular Bcr-Abl kinase activity that target the myristate binding site. Here, we used cell-based structure−activity relationships to guide the optimization and diversification of ligands that are capable of binding to the myristate binding site and rationalize the findings based upon an Abl−compound 1 cocrystal. We elucidate the structure−activity relationships required to obtain potent antiproliferative activity against Bcr-Abl transformed cells and report the discovery of new compounds (5g, 5h, 6a, 14d, and 21j-I) that display improved potency or pharmacological properties. This work demonstrates that a variety of structures can effectively target the Bcr-Abl myristate binding site and provides new leads for developing drugs that can target this binding site.


Blood | 2014

PIM inhibitors target CD25-positive AML cells through concomitant suppression of STAT5 activation and degradation of MYC oncogene

Zhuyan Guo; Anlai Wang; Weidong Zhang; Mikhail Levit; Qiang Gao; Claude Barberis; Michel Tabart; Jingxin Zhang; Dietmar Hoffmann; Dmitri Wiederschain; Jennifer Rocnik; Fangxian Sun; Josh Murtie; Christoph Lengauer; Stefan Gross; Bailin Zhang; Hong Cheng; Vinod F. Patel; Laurent Schio; Francisco Adrian; Marion Dorsch; Carlos Garcia-Echeverria; Shih Min A Huang

Postchemotherapy relapse presents a major unmet medical need in acute myeloid leukemia (AML), where treatment options are limited. CD25 is a leukemic stem cell marker and a conspicuous prognostic marker for overall/relapse-free survival in AML. Rare occurrence of genetic alterations among PIM family members imposes a substantial hurdle in formulating a compelling patient stratification strategy for the clinical development of selective PIM inhibitors in cancer. Here we show that CD25, a bona fide STAT5 regulated gene, is a mechanistically relevant predictive biomarker for sensitivity to PIM kinase inhibitors. Alone or in combination with tyrosine kinase inhibitors, PIM inhibitors can suppress STAT5 activation and significantly shorten the half-life of MYC to achieve substantial growth inhibition of high CD25-expressing AML cells. Our results highlight the importance of STAT5 and MYC in rendering cancer cells sensitive to PIM inhibitors. Because the presence of a CD25-positive subpopulation in leukemic blasts correlates with poor overall or relapse-free survival, our data suggest that a combination of PIM inhibitors with chemotherapy and tyrosine kinase inhibitors could improve long-term therapeutic outcomes in CD25-positive AML.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of novel 1H-imidazol-2-yl-pyrimidine-4,6-diamines as potential antimalarials.

Xianming Deng; Advait Nagle; Tao Wu; Tomoyo Sakata; Kerstin Henson; Zhong Chen; Kelli Kuhen; David Plouffe; Elizabeth A. Winzeler; Francisco Adrian; Tove Tuntland; Jonathan Chang; Susan Simerson; Steven Howard; Jared Ek; John Isbell; David C. Tully; Arnab K. Chatterjee; Nathanael S. Gray

A novel family of 1H-imidazol-2-yl-pyrimidine-4,6-diamines has been identified with potent activity against the erythrocyte-stage of Plasmodium falciparum (Pf), the most common causative agent of malaria. A systematic SAR study resulted in the identification of compound 40 which exhibits good potency against both wild-type and drug resistant parasites and exhibits good in vivo pharmacokinetic properties.


ACS Medicinal Chemistry Letters | 2015

(R)-2-Phenylpyrrolidine Substituted Imidazopyridazines: A New Class of Potent and Selective Pan-TRK Inhibitors.

Ha-Soon Choi; Paul Vincent Rucker; Zhicheng Wang; Yi Fan; Pamela A. Albaugh; Greg Chopiuk; Francois Gessier; Fangxian Sun; Francisco Adrian; Guoxun Liu; Tami Hood; Nanxin Li; Yong Jia; Jianwei Che; Susan McCormack; Allen Li; Jie Li; Auzon Steffy; AnneMarie Culazzo; Celine Tompkins; Van Phung; Andreas Kreusch; Min Lu; Bin Hu; Apurva Chaudhary; Mahavir Prashad; Tove Tuntland; Bo Liu; Jennifer L. Harris; H. Martin Seidel

Deregulated kinase activities of tropomyosin receptor kinase (TRK) family members have been shown to be associated with tumorigenesis and poor prognosis in a variety of cancer types. In particular, several chromosomal rearrangements involving TRKA have been reported in colorectal, papillary thyroid, glioblastoma, melanoma, and lung tissue that are believed to be the key oncogenic driver in these tumors. By screening the Novartis compound collection, a novel imidazopyridazine TRK inhibitor was identified that served as a launching point for drug optimization. Structure guided drug design led to the identification of (R)-2-phenylpyrrolidine substituted imidazopyridazines as a series of potent, selective, orally bioavailable pan-TRK inhibitors achieving tumor regression in rats bearing KM12 xenografts. From this work the (R)-2-phenylpyrrolidine has emerged as an ideal moiety to incorporate in bicyclic TRK inhibitors by virtue of its shape complementarity to the hydrophobic pocket of TRKs.

Collaboration


Dive into the Francisco Adrian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Taebo Sim

Korea Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tove Tuntland

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge