Network


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

Hotspot


Dive into the research topics where Siavosh Mahboobi is active.

Publication


Featured researches published by Siavosh Mahboobi.


Journal of Medicinal Chemistry | 2001

Synthetic 2-aroylindole derivatives as a new class of potent tubulin-inhibitory, antimitotic agents.

Siavosh Mahboobi; Herwig Pongratz; Harald Hufsky; Jörg Hockemeyer; Markus Frieser; Alexei Lyssenko; D. H. Paper; Jutta Bürgermeister; Frank-D. Böhmer; Heinz-Herbert Fiebig; Angelika M. Burger; Silke Baasner; Thomas Beckers

A new class of simple synthetic antimitotic compounds based on 2-aroylindoles was discovered. (5-Methoxy-1H-2-indolyl)-phenylmethanone (1) as well as analogous 3-fluorophenyl- (36) and 3-methoxyphenyl (3) derivatives displayed high cytotoxicity of IC(50) = 20 to 75 nM against the human HeLa/KB cervical, SK-OV-3 ovarian, and U373 astrocytoma carcinoma cell lines. The inhibition of proliferation correlated with the arrest in the G2/M phase of the cell cycle. In in vitro assays with tubulin isolated from bovine brain, in general antiproliferative activity correlated with inhibition of tubulin polymerization. Thus, the antimitotic activity of 2-aroylindoles is explained by interference with the mitotic spindle apparatus and destabilization of microtubules. In contrast to colchicine, vincristine, nocodazole, or taxol, 1 did not significantly affect the GTPase activity of beta-tubulin. Interestingly, selected compounds inhibited angiogenesis in the chorioallantoic membrane (CAM) assay. In xenograft experiments, 1 was highly active after oral administration at 200 mg/kg against the human amelanocytic melanoma MEXF 989 in athymic nude mice. We conclude, that 2-aroylindoles constitute an interesting new class of antitubulin agents with the potential to be clinically developed for cancer treatment.


Journal of Biological Chemistry | 2003

A Single Amino Acid Exchange Inverts Susceptibility of Related Receptor Tyrosine Kinases for the ATP Site Inhibitor STI-571

Frank-D. Böhmer; Luchezar Karagyozov; Andrea Uecker; Hubert Serve; Alexander Botzki; Siavosh Mahboobi; Stefan Dove

The tyrosine kinase inhibitor STI-571 potently blocks BCR-Abl, platelet-derived growth factor (PDGF) α- and β-receptors, and c-Kit kinase activity. Flt3, a receptor tyrosine kinase closely related to PDGF receptors and c-Kit is, however, not inhibited by STI-571. Sequence alignments of different kinases and indications from the crystal structure of the STI-571 Abl kinase complex revealed amino acid residues that are probably crucial for this activity profile. It was predicted that Flt3 Phe-691 in the β5 strand may sterically prevent interaction with STI-571. The point mutants Flt3 F691T and PDGFβ-receptor T681F were constructed, and kinase assays showed that the Flt3 mutant but not the PDGFβ-receptor mutant is inhibited by STI-571. Docking of STI-571 into computer models of the PDGFβ-receptor and Flt3 kinase domains and comparison with the crystal structure of the STI-571 Abl kinase complex indicated very similar binding sites among the three nonphosphorylated kinases, suggesting corresponding courses of their Asp-Phe-Gly motifs and activation loops. Accordingly, we observed reduced sensitivity of preactivated compared with nonactivated PDGFR-β for the inhibition by STI-571. Courses of the activation loop that collide with STI-571 binding explain its inactivity at other kinases as the insulin receptor. The binding site models of PDGFR-β and Flt3 were applied to predict structural approaches for more selective PDGFβ-receptor inhibitors.


Journal of Medicinal Chemistry | 2009

Design of chimeric histone deacetylase- and tyrosine kinase-inhibitors: a series of imatinib hybrides as potent inhibitors of wild-type and mutant BCR-ABL, PDGF-Rbeta, and histone deacetylases.

Siavosh Mahboobi; Stefan Dove; Andreas Sellmer; Matthias Winkler; Emerich Eichhorn; Herwig Pongratz; Thomas Ciossek; Thomas Baer; Thomas Maier; Thomas Beckers

Inhibitors of histone deacetylases are a new class of cancer therapeutics with possibly broad applicability. Combinations of HDAC inhibitors with the kinase inhibitor 1 (Imatinib) in recent studies showed additive and synergistic effects. Here we present a new concept by combining inhibition of protein kinases and HDACs, two independent pharmacological activities, in one synthetic small molecule. In general, the HDAC inhibition profile, the potencies, and the probable binding modes to HDAC1 and HDAC6 were similar as for 6 (SAHA). Inhibition of Abl kinase in biochemical assays was maintained for most compounds, but in general the kinase selectivity profile differed from that of 1 with nearly equipotent inhibition of the wild-type and the Imatinib resistant Abl T(315)I mutant. A potent cellular inhibition of PDGFR and cytotoxicity toward EOL-1 cells, a model for idiopathic hypereosinophilic syndrome (HES), are restored or enhanced for selected analogues (12b, 14b, and 18b). Cytotoxicity was evaluated by using a broad panel of tumor cell lines, with selected analogues displaying mean IC(50) values between 3.6 and 7.1 muM.


Journal of Medicinal Chemistry | 2010

Novel chimeric histone deacetylase inhibitors: a series of lapatinib hybrides as potent inhibitors of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and histone deacetylase activity.

Siavosh Mahboobi; Andreas Sellmer; Matthias Winkler; Emerich Eichhorn; Herwig Pongratz; Thomas Ciossek; Thomas Baer; Thomas Maier; Thomas Beckers

Reversible lysine-specific acetylation has been described as an important posttranslational modification, regulating chromatin structure and transcriptional activity in the case of core histone proteins. Histone deacetylases (HDAC) are considered as a promising target for anticancer drug development, with 2a as pan-HDAC inhibitor approved for cutanous T-cell lymphoma therapy and several other HDAC inhibitors currently in preclinical and clinical development. Protein kinases are a well-established target for cancer therapy with the EGFR/HER2 inhibitor 5 approved for treatment of advanced, HER2 positive breast cancer as a prominent example. In the present report, we present a novel strategy for cancer drug development by combination of EGFR/HER2 kinase and HDAC inhibitory activity in one molecule. By combining the structural features of 5 with an (E)-3-(aryl)-N-hydroxyacrylamide motif known from HDAC inhibitors like 1 or 3, we obtained selective inhibitors for both targets with potent cellular activity (target inhibition and cytotoxicity) of selected compounds 6a and 6c. By combining two distinct pharmacologically properties in one molecule, we postulate a broader activity spectrum and less likelihood of drug resistance in cancer patients.


Trends in Pharmacological Sciences | 2014

Drugging the HDAC6–HSP90 interplay in malignant cells

Oliver H. Krämer; Siavosh Mahboobi; Andreas Sellmer

Acetylation and deacetylation cycles regulate crucial biological processes. The enzymes deacetylating lysine residues are termed histone deacetylases (HDACs). Eighteen deacetylases have been isolated from mammalian cells. There is an intense search underway for individual functions of these enzymes and for selective histone deacetylase inhibitors (HDACi). HDAC6 in particular has unique cytoprotective functions that rely on its ability to ensure protein homeostasis and to prevent protein aggregation. The chaperone heat shock protein 90 (HSP90) also safeguards proteins and is deacetylated by HDAC6. Current data illustrate the complexity and importance of the HDAC6-HSP90 interplay. In this review, we discuss how recently identified HSP90-dependent regulators of posttranslational modifications (PTMs) of HDAC6 dictate its functions, and how HDACi-induced acetylation of HSP90 might control oncologically relevant proteins, especially in leukemic cells. Additionally, we discuss small molecules blocking HDAC6 and how such agents could become therapeutically relevant. We summarize structure-function relationships that determine the specificity of drugs against HDAC6.


Journal of Medicinal Chemistry | 2009

2-Arylamino-4-Amino-5-Aroylthiazoles. "One-Pot" Synthesis and Biological Evaluation of a New Class of Inhibitors of Tubulin Polymerization

Romeo Romagnoli; Pier Giovanni Baraldi; Maria Dora Carrion; Olga Cruz-Lopez; Carlota Lopez Cara; Giuseppe Basso; Giampietro Viola; Mohammed Khedr; Jan Balzarini; Siavosh Mahboobi; Andreas Sellmer; Andrea Brancale; Ernest Hamel

The essential role of microtubules in mitosis makes them a major target of compounds useful for cancer therapy. In our search for potent antitumor agents, a novel series of 2-anilino-4-amino-5-aroylthiazoles was synthesized and evaluated for antiproliferative activity, inhibition of tubulin polymerization, and cell cycle effects. SAR was elucidated with various substitutions on the phenylamino and aroyl moiety at the 2- and 5-positions, respectively, of the 4-aminothiazole skeleton. Tumor cell exposure to several of these compounds led to the arrest of HeLa cells in the G2/M phase of the cell cycle and induction of apoptosis.


Bioorganic & Medicinal Chemistry | 2012

Novel inhibitors of epidermal growth factor receptor: (4-(Arylamino)-7H-pyrrolo[2,3-d]pyrimidin-6-yl)(1H-indol-2-yl)methanones and (1H-indol-2-yl)(4-(phenylamino)thieno[2,3-d]pyrimidin-6-yl)methanones.

Thomas Beckers; Andreas Sellmer; Emerich Eichhorn; Herwig Pongratz; Christoph Schächtele; Frank Totzke; Gerhard Kelter; Rebekka Krumbach; Heinz-Herbert Fiebig; Frank-D. Böhmer; Siavosh Mahboobi

Several members of the quinazoline class of known tyrosine kinase inhibitors are approved anticancer agents, often showing selectivity for receptors of the HER/ErbB-family. Combining structural elements of this class with the bisindolylmethanone-structure led to a series of novel compounds. These compounds inhibited EGFR in the nanomolar range. Moreover, inhibition of EGFR autophosphorylation in intact A431 cells was shown, with IC(50) values ranging form 0.3-1μM for compound 42, and 0.1-0.3μM for 45. In a panel of 42 human tumor cell lines the sensitivity profile of the novel compounds was shown to be similar to that of the quinazoline class of tyrosine kinase inhibitors lapatinib and erlotinib (Tarceva®).


British Journal of Haematology | 2009

Bis(1H‐indol‐2‐yl)methanones are effective inhibitors of FLT3‐ITD tyrosine kinase and partially overcome resistance to PKC412A in vitro

Florian H. Heidel; Daniel B. Lipka; Fian K. Mirea; Siavosh Mahboobi; Rebekka Grundler; Rama Krishna Kancha; Justus Duyster; Michael Naumann; Christoph Huber; Frank-D. Böhmer; Thomas Fischer

Inhibition of the mutated fms‐like tyrosine kinase 3 (FLT3) receptor tyrosine kinase is a promising therapeutic strategy in acute myeloid leukaemia (AML). However, development of resistance to FLT3 tyrosine kinase inhibitors (TKI), such as PKC412A, has been described recently. This observation may have an increasing impact on the duration of response and relapse rates in upcoming clinical trials employing FLT3‐TKI. Herein we investigated two representatives of a novel class of FLT3‐TKI: Bis(1H‐indol‐2‐yl)methanones. Both compounds effectively induced apoptosis in FLT3‐internal tandem duplicate (ITD)‐transfected murine myeloid cells and in primary FLT3‐ITD positive blasts. Combination of both compounds with chemotherapy revealed synergistic effects in apoptosis assays. The compounds did not show significant toxicity in human bone marrow cells derived from healthy donors. Compound102 overcame resistance to PKC412 within a non‐myelotoxic dose‐range. Western Blotting experiments of 32D‐FLT3‐ITD cells showed dose‐dependent dephosphorylation of FLT3‐ITD and of its downstream targets STAT5, AKT and ERK upon incubation with either compound. In conclusion, bis(1H‐indol‐2‐yl)methanones overcome resistance mediated by FLT3‐ITD mutations at position N676 and show strong efficacy in FLT3‐ITD‐positive cells alone as well as in combination with chemotherapy. We propose that further development of methanone compounds overcoming resistance to currently established FLT3‐TKIs is an important step forward to an anticipated need within our future therapeutic algorithm in FLT3‐ITD‐positive AML.


Biochemical Journal | 2004

The tyrosine kinase FRK/RAK participates in cytokine-induced islet cell cytotoxicity

Michael Welsh; Charlotte Welsh; Maria Ekman; Johan Dixelius; Robert Hägerkvist; Cecilia Annerén; Björn Åkerblom; Siavosh Mahboobi; Subhashini Chandrasekharan; Edison T. Liu

Hallmarks of the inflammatory process in Type I diabetes are macrophage activation, local release of beta-cell-toxic cytokines and infiltration of cytotoxic T lymphocytes. We have observed recently that mice overexpressing active FRK (fyn-related kinase)/RAK (previously named GTK/Bsk/IYK, where GTK stands for gut tyrosine kinase, Bsk for beta-cell Src-homology kinase and IYK for intestinal tyrosine kinase) in beta-cells exhibit increased susceptibility to beta-cell-toxic events, and therefore, we now attempt to find a more precise role for FRK/RAK in these processes. Phosphopeptide mapping of baculovirus-produced mouse FRK/RAK revealed an autophosphorylation pattern compatible with Tyr-394 being the main site. No evidence for in vitro phosphorylation of the C-terminal regulatory sites Tyr-497 and Tyr-504 was obtained, nor was there any indication of in vitro regulation of FRK/RAK kinase activity. Screening a panel of known tyrosine kinase inhibitors for their ability to inhibit FRK/RAK revealed several compounds that inhibited FRK/RAK, with a potency similar to that reported for their ability to inhibit other tyrosine kinases. Cytokine-induced islet toxicity was reduced in islets isolated from FRK/RAK knockout mice and this occurred without effects on the production of nitric oxide. Addition of the nitric oxide inhibitor nitroarginine to FRK/RAK knockout islets exposed to cytokines decreased cell death to a basal level. In normal islets, cytokine-induced cell death was inhibited by the addition of two FRK/RAK inhibitors, SU4984 and D-65495, or by transfection with short interfering RNA against FRK/RAK. It is concluded that FRK/RAK contributes to cytokine-induced beta-cell death, and inhibition of this kinase could provide means to suppress beta-cell destruction in Type I diabetes.


Cellular Signalling | 2017

Class I histone deacetylases regulate p53/NF-κB crosstalk in cancer cells

Claudia Schäfer; Anja Göder; Mandy Beyer; Nicole Kiweler; Nisintha Mahendrarajah; Anke Rauch; Teodora Nikolova; Natasa Stojanovic; Martin Wieczorek; Thomas R. Reich; Maja T. Tomicic; Jürgen Sonnemann; Sascha Dietrich; Andreas Sellmer; Siavosh Mahboobi; Thorsten Heinzel; Günter Schneider; Oliver H. Krämer

The transcription factors NF-κB and p53 as well as their crosstalk determine the fate of tumor cells upon therapeutic interventions. Replicative stress and cytokines promote signaling cascades that lead to the co-regulation of p53 and NF-κB. Consequently, nuclear p53/NF-κB signaling complexes activate NF-κB-dependent survival genes. The 18 histone deacetylases (HDACs) are epigenetic modulators that fall into four classes (I-IV). Inhibitors of histone deacetylases (HDACi) become increasingly appreciated as anti-cancer agents. Based on their effects on p53 and NF-κB, we addressed whether clinically relevant HDACi affect the NF-κB/p53 crosstalk. The chemotherapeutics hydroxyurea, etoposide, and fludarabine halt cell cycle progression, induce DNA damage, and lead to DNA fragmentation. These agents co-induce p53 and NF-κB-dependent gene expression in cell lines from breast and colon cancer and in primary chronic lymphatic leukemia (CLL) cells. Using specific HDACi, we find that the class I subgroup of HDACs, but not the class IIb deacetylase HDAC6, are required for the hydroxyurea-induced crosstalk between p53 and NF-κB. HDACi decrease the basal and stress-induced expression of p53 and block NF-κB-regulated gene expression. We further show that class I HDACi induce senescence in pancreatic cancer cells with mutant p53.

Collaboration


Dive into the Siavosh Mahboobi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Beckers

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan Dove

University of Regensburg

View shared research outputs
Top Co-Authors

Avatar

Alfred Popp

University of Regensburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge