Network


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

Hotspot


Dive into the research topics where Hendrik Falk is active.

Publication


Featured researches published by Hendrik Falk.


Nature Communications | 2015

A RIPK2 inhibitor delays NOD signalling events yet prevents inflammatory cytokine production

Ueli Nachbur; Che A. Stafford; Aleksandra Bankovacki; Yifan Zhan; Lisa Lindqvist; Berthe Katrine Fiil; Yelena Khakham; Hyun Ja Ko; Jarrod J. Sandow; Hendrik Falk; Jessica K. Holien; Diep Chau; Joanne M. Hildebrand; James E. Vince; Phillip P Sharp; Andrew I. Webb; Katherine A. Jackman; Sabrina Mühlen; Catherine L. Kennedy; Kym N. Lowes; James M. Murphy; Mads Gyrd-Hansen; Michael W. Parker; Elizabeth L. Hartland; Andrew M. Lew; David C. S. Huang; Guillaume Lessene; John Silke

Intracellular nucleotide binding and oligomerization domain (NOD) receptors recognize antigens including bacterial peptidoglycans and initiate immune responses by triggering the production of pro-inflammatory cytokines through activating NF-κB and MAP kinases. Receptor interacting protein kinase 2 (RIPK2) is critical for NOD-mediated NF-κB activation and cytokine production. Here we develop and characterize a selective RIPK2 kinase inhibitor, WEHI-345, which delays RIPK2 ubiquitylation and NF-κB activation downstream of NOD engagement. Despite only delaying NF-κB activation on NOD stimulation, WEHI-345 prevents cytokine production in vitro and in vivo and ameliorates experimental autoimmune encephalomyelitis in mice. Our study highlights the importance of the kinase activity of RIPK2 for proper immune responses and demonstrates the therapeutic potential of inhibiting RIPK2 in NOD-driven inflammatory diseases.


Journal of Proteome Research | 2013

Characterization of the Novel Broad-Spectrum Kinase Inhibitor CTx-0294885 As an Affinity Reagent for Mass Spectrometry-Based Kinome Profiling

Luxi Zhang; Ian Peter Holmes; Falko Hochgräfe; Scott Raymond Walker; Naveid Ali; Emily S. Humphrey; Jianmin Wu; Melanie de Silva; Wilhelmus J A Kersten; Theresa Connor; Hendrik Falk; Lynda Allan; Ian P. Street; John D. Bentley; Patricia A. Pilling; Brendon J. Monahan; Thomas S. Peat; Roger J. Daly

Kinase enrichment utilizing broad-spectrum kinase inhibitors enables the identification of large proportions of the expressed kinome by mass spectrometry. However, the existing inhibitors are still inadequate in covering the entire kinome. Here, we identified a novel bisanilino pyrimidine, CTx-0294885, exhibiting inhibitory activity against a broad range of kinases in vitro, and further developed it into a Sepharose-supported kinase capture reagent. Use of a quantitative proteomics approach confirmed the selectivity of CTx-0294885-bound beads for kinase enrichment. Large-scale CTx-0294885-based affinity purification followed by LC-MS/MS led to the identification of 235 protein kinases from MDA-MB-231 cells, including all members of the AKT family that had not been previously detected by other broad-spectrum kinase inhibitors. Addition of CTx-0294885 to a mixture of three kinase inhibitors commonly used for kinase-enrichment increased the number of kinase identifications to 261, representing the largest kinome coverage from a single cell line reported to date. Coupling phosphopeptide enrichment with affinity purification using the four inhibitors enabled the identification of 799 high-confidence phosphosites on 183 kinases, ∼10% of which were localized to the activation loop, and included previously unreported phosphosites on BMP2K, MELK, HIPK2, and PRKDC. Therefore, CTx-0294885 represents a powerful new reagent for analysis of kinome signaling networks that may facilitate development of targeted therapeutic strategies. Proteomics data have been deposited to the ProteomeXchange Consortium ( http://proteomecentral.proteomexchange.org ) via the PRIDE partner repository with the data set identifier PXD000239.


Journal of Biomolecular Screening | 2011

An Efficient High-Throughput Screening Method for MYST Family Acetyltransferases, a New Class of Epigenetic Drug Targets

Hendrik Falk; Theresa Connor; Hong Yang; Karen J. Loft; Joanne L. Alcindor; George Nikolakopoulos; Regina Surjadi; John D. Bentley; Meghan Hattarki; Olan Dolezal; James M. Murphy; Brendon J. Monahan; Thomas S. Peat; Tim Thomas; Jonathan B. Baell; John P. Parisot; Ian P. Street

Epigenetic aberrations are increasingly regarded as key factors in cancer progression. Recently, deregulation of histone acetyltransferases (HATs) has been linked to several types of cancer. Monocytic leukemia zinc finger protein (MOZ) is a member of the MYST family of HATs, which regulate gene expression in cell proliferation and differentiation. Deregulation of these processes through constitutively active MOZ fusion proteins gives rise to the formation of leukemic stem cells, rendering MOZ an excellent target for treating myeloid leukemia. The authors implemented a hit discovery campaign to identify small-molecule inhibitors of MOZ-HAT activity. They developed a robust, homogeneous assay measuring the acetylation of synthetic histone peptides. In a primary screening campaign testing 243 000 lead-like compounds, they identified inhibitors from several chemical classes. Secondary assays were used to eliminate assay-interfering compounds and prioritize confirmed hits. This study establishes a new high-throughput assay for HAT activity and could provide the foundation for the development of a new class of drugs for the treatment of leukemias.


Bioorganic & Medicinal Chemistry Letters | 2011

Identification of 5,6-substituted 4-aminothieno[2,3-d]pyrimidines as LIMK1 inhibitors.

Brad E. Sleebs; George Nikolakopoulos; Ian P. Street; Hendrik Falk; Jonathan B. Baell

4-Aminobenzothieno[3,2-d]pyrimidines were previously identified in a high throughput screening campaign as LIMK1 inhibitors. Scaffold reversal led to the identification of a series of simple 5,6-substituted 4-aminothieno[2,3-d]pyrimidines with low micromolar inhibition of LIMK1.


MedChemComm | 2011

Identification of 3-aminothieno[2,3-b]pyridine-2-carboxamides and 4-aminobenzothieno[3,2-d]pyrimidines as LIMK1 inhibitors

Brad E. Sleebs; Alla Levit; Ian P. Street; Hendrik Falk; Tim Hammonds; Ai Ching Wong; Mark Charles; Michael F. Olson; Jonathan B. Baell

A high throughput chemical screening campaign has led to the identification of 3-aminobenzo[b]thiophene-2-carboxamides as LIMK1 inhibitors. Evolution of bicyclic hits to the tricyclic 4-aminobenzothieno[3,2-d]pyrimidine, using a traditional medicinal chemistry SAR guided approach, resulted in a significant increase in potency. Further elaboration has seen the 7-phenyl-4-aminobenzothieno[3,2-d]pyrimidine emerge as a LIMK1 inhibitor lead candidate.


MedChemComm | 2011

Development of substituted 7-phenyl-4-aminobenzothieno[3,2-d] pyrimidines as potent LIMK1 inhibitors

Brad E. Sleebs; Danny Ganame; Alla Levit; Ian P. Street; Alison Gregg; Hendrik Falk; Jonathan B. Baell

7-Phenyl-4-aminobenzothieno[3,2-d]pyrimidines were previously reported to exhibit moderate LIMK1 inhibition. Further exploration of SAR around the 7-phenyl moiety has led to the development of a lead series with an increased potency for LIMK1. Evaluation of physicochemical and ADME properties, and off-target kinase screens has seen this novel series emerge as a promising platform for a set of tool compounds for evaluating LIMK as a therapeutic target.


Combinatorial Chemistry & High Throughput Screening | 2014

A Perspective on 10-Years HTS Experience at the Walter and Eliza Hall Institute of Medical Research – Eighteen Million Assays and Counting

Kurt Lackovic; Guillaume Lessene; Hendrik Falk; Karl-Johan Leuchowius; Jonathan B. Baell; Ian P. Street

The Walter and Eliza Hall Institute of Medical Research (WEHI) is Australias longest serving medical research institute. WEHIs High Throughput Screening (HTS) Facility was established in 2003 with


Cancer Research | 2015

Abstract 5371: PRMT5 inhibitors as novel treatment for cancers

Hendrik Falk; Richard Foitzik; Elizabeth Allan; Melanie deSilva; Hong Yang; Ylva E. Bozikis; Marica Nikac; Scott Raymond Walker; Michelle A. Camerino; Ben J. Morrow; Alexandra Elizabeth Stupple; Rachel Lagiakos; Jo-Anne Pinson; Romina Lessene; Wilhelmus J A Kersten; Danny Ganame; Ian Peter Holmes; Gill E. Lunniss; Matthew Chung; Stefan J. Hermans; Michael W. Parker; Alison Thistlethwaite; Karen L. White; Susan A. Charman; Brendon J. Monahan; Patricia A. Pilling; Julian Grusovin; Thomas S. Peat; Stefan Eugen Sonderegger; Emma Toulmin

5 million of infrastructure funds invested by WEHI, and the Victorian State Governments Strategic Technology Initiative through Bio21 Australia Ltd. The Facility was Australias first truly academic HTS facility and was one of only a handful operating in publicly funded institutions worldwide at that time. The objectives were to provide access to enabling HTS technologies, such as assay design, liquid handling automation, compound libraries and expertise to promote translation of basic research in a national setting that has a relatively young biotech sector and does not have a big Pharma research presence. Ten years on and the WEHI HTS Facility has participated in over 92 collaborative projects, generated over 18 million data points, and most importantly, projects that began in the Facility have been commercialized successfully (due to strong ties with Business Development and emphasis on intellectual property management) and now have molecules progressing in clinical trials.


Cancer Research | 2014

Abstract 4029: BL-011256 is a novel VEGFR3 selective inhibitor, which suppresses tumor lymphatics and lymph node metastasis in an animal model of melanoma

Annabell F. Leske; Richard Foitzik; Donna M. Beaumont; John D. Bentley; Ylva Bergman; Chloe K. Brown; Michelle A. Camerino; Susan A. Charman; Neil Choi; Melanie D. Silva; Matthew Chung; Hendrik Falk; Danny Ganame; Alison Gregg; Julian Grusovin; Andrew John Harvey; Catherine Fae Hemley; Ian Peter Holmes; Belinda Huff; Daniel J. Inglis; Wilhelmus J A Kersten; Tina C. Lavranos; Romina Lessene; Gillian Elizabeth Lunniss; Brendon J. Monahan; Benjamin Joseph Morrow; Marica Nikac; George Nikolakopoulos; Dharam Paul; Tom Peat

Increased expression or dysregulation of protein arginine methyltransferase 5 (PRMT5) activity is associated with poor prognosis in many cancers. Through increased methylation of epigenetic and non-epigenetic targets, the aberrant activity of PRMT5 has been associated with many pro-tumourigenic cellular changes such as, increased levels of protein synthesis, dysregulation of cell cycle, cellular adaptation to hypoxic conditions, and suppression of normal cell death pathways. Genetic studies suggest that suppression of PRMT5 activity can reverse many of these pro-tumourigenic effects making PRMT5 an attractive drug discovery target. We screened a library of 350,000 lead-like compounds with a biochemical assay measuring the methylation of a histone H4 peptide by the recombinant human PRMT5/MEP50 complex. Biochemical and biophysical profiling of the inhibitory compounds indicated that several distinct binding modes were exhibited by the different chemical scaffolds. Inhibitors displayed competitive, noncompetitive or uncompetitive interactions with respect to S-adenosyl methionine and the peptide substrate. Medicinal chemistry developed several classes of potent, highly selective inhibitors of PRMT5 methyltransferase activity from the hit set. The optimised tool compound, CTx-034, is a potent inhibitor of PRMT5 methyl transferase activity (KD = 2 nM), which is highly selective (>100-fold) versus a panel of 18 methyltransferases (including 6 PRMT family members), 11 lysine demethylases, and 15 safety related targets (GPCRs, ion channels, enzymes). Treatment of cancer cell lines with CTx-034 reduces cellular levels of symmetrically dimethylated H4 Arginine 3 (H4R3me2s), in a dose dependent manner (IC50 = 4 nM) to levels undetectable by Western blot. Furthermore, within this chemical series the ability of compounds to reduce cellular levels of H4R3me2s closely correlates with PRMT5 inhibitory activity supporting PRMT5 as the cellular target of these compounds, and suggesting that PRMT5 is the major writer of this histone mark in many cancer cell lines. CTx-034 also inhibits the symmetric dimethylation of arginine on other histone and non-histone cellular substrates of PRMT5, including H3R2me2s and SmD1. Conversely, CTx-034 treatment does not reduce levels of H4R3 asymmetric dimethylation, a histone mark catalysed by PRMT1. Finally, CTx-034 has good oral bioavailability and pharmacokinetic properties in rodents and twice-daily dosing (10 - 100 mg/kg) over 10-14 days produces a dose dependent reduction of the H4R3me2s mark in bone marrow cells and peripheral white blood cells. This treatment is well tolerated by the mice, with no significant reduction in body weight or changes in haematological parameters observed. CTx-034 provides an excellent tool compound for cellular and in vivo proof of concept studies. Citation Format: Hendrik Falk, Richard C. Foitzik, Elizabeth Allan, Melanie deSilva, Hong Yang, Ylva E. Bozikis, Marica Nikac, Scott R. Walker, Michelle A. Camerino, Ben J. Morrow, Alexandra E. Stupple, Rachel Lagiakos, Jo-Anne Pinson, Romina Lessene, Wilhelmus JA Kersten, Danny G. Ganame, Ian P. Holmes, Gill E. Lunniss, Matthew Chung, Stefan J. Hermans, Michael W. Parker, Alison Thistlethwaite, Karen White, Susan A. Charman, Brendon J. Monahan, Patricia Pilling, Julian Grusovin, Thomas S. Peat, Stefan Sonderegger, Emma Toulmin, Stephen M. Jane, David J. Curtis, Paul A. Stupple, Ian P. Street. PRMT5 inhibitors as novel treatment for cancers. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5371. doi:10.1158/1538-7445.AM2015-5371


Nature | 2018

Inhibitors of histone acetyltransferases KAT6A/B induce senescence and arrest tumour growth

Jonathan B. Baell; D.J. Leaver; Stefan J. Hermans; Gemma L. Kelly; Brennan; Natalie L. Downer; Nhu-Y Nguyen; J. Wichmann; Helen M. McRae; Ying Ting Yang; B. Cleary; H.R. Lagiakos; S. Mieruszynski; G. Pacini; Hannah K. Vanyai; M.I. Bergamasco; R.E. May; B.K. Davey; K.J. Morgan; A.J. Sealey; Bin Wang; N. Zamudio; Stephen Wilcox; Alexandra L. Garnham; Bilal N. Sheikh; Brandon J. Aubrey; Karen Doggett; M.C. Chung; M. de Silva; John D. Bentley

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The role of VEGFR3 in lymphangiogenesis has been well established. Targeting VEGFR3 has been shown to curtail tumor progression mediated via lymphatic dissemination. More recently VEGFR3 was shown to play an important role in the mediation of tumor-induced immune cell tolerance. We have identified BL-011256, a novel inhibitor of VEGFR3 that exhibits 17-fold selectivity over VEGFR2 and a narrow tyrosine kinase inhibition spectrum. A 7-day b.i.d repeat oral dose study showed that BL-011256 is well tolerated in mice. Mouse plasma exposure experiments demonstrated that BL-011256 attains free drug plasma concentration levels that exceed the concentration required for IC50 activity on VEGFR3 in vitro but are considerably lower than the in vitro IC50 required for activity on VEGFR2. In the B16F10 mouse melanoma model, animals bearing melanoma tumors displayed considerably attenuated signs of tumor progression when treated with BL-011256. BL-011256 caused a 70% reduction in primary lesion growth and a 50% reduction in metastasis to the draining lymph node. Furthermore, BL-011256 was active in reducing the number of satellite in-transit metastases. Immunohistochemical whole mount analyses on ears with primary tumor lesions derived from BL-011256 treated and vehicle-treated mice using Lyve-1 for the identification of lymphatic vessels and CD31 for the identification of blood vessels was conducted. Tumors in vehicle-treated mice displayed a peri-tumoral area densely populated by lymphatic vessels. In contrast, tumors derived from BL-011256 treated mice were devoid of peri-tumoral lymphatic vessels. Notably, both vehicle-treated and BL-011256-treated animals displayed similar staining for peritumoral blood vessels, suggesting no effect on blood vessels (this is consistent with no activity on VEGFR2). Furthermore, PK sampling during the last day of dosing in a 14-day dosing schedule demonstrated that there is no compound accumulation during the repeat dosing schedule utilised in the B16F10 tumour efficacy experiment. In conclusion BL-011256 has been identified as a selective inhibitor of VEGFR3 that supresses both primary tumor growth and lymph node metastasis. Citation Format: Annabell Leske, Richard Foitzik, Donna Beaumont, John Bentley, Ylva Bergman, Chloe Brown, Michelle Camerino, Susan Charman, Neil Choi, Melanie De Silva, Matthew Chung, Hendrik Falk, Danny Ganame, Alison Gregg, Julian Grusovin, Andrew Harvey, Catherine Hemley, Ian Holmes, Belinda Huff, Daniel Inglis, Wilhelmus Kersten, Tina Lavranos, Romina Lessene, Gillian Lunniss, Brendon Monahan, Benjamin Morrow, Marica Nikac, George Nikolakopoulos, Dharam Paul, Tom Peat, Justin Ripper, Michaela Scherer, Paul Stupple, Karen White, Ian Street, Gabriel Kremmidiotis. BL-011256 is a novel VEGFR3 selective inhibitor, which suppresses tumor lymphatics and lymph node metastasis in an animal model of melanoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4029. doi:10.1158/1538-7445.AM2014-4029

Collaboration


Dive into the Hendrik Falk's collaboration.

Top Co-Authors

Avatar

Ian P. Street

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brendon J. Monahan

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar

Brad E. Sleebs

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

George Nikolakopoulos

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ian Peter Holmes

Cooperative Research Centre

View shared research outputs
Top Co-Authors

Avatar

John D. Bentley

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danny Ganame

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge