Network


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

Hotspot


Dive into the research topics where Gali Golan is active.

Publication


Featured researches published by Gali Golan.


Nature Communications | 2017

A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action

Elena Campaner; Alessandra Rustighi; Alessandro Zannini; Alberto Cristiani; Silvano Piazza; Yari Ciani; Ori Kalid; Gali Golan; Erkan Baloglu; Sharon Shacham; Barbara Valsasina; Ulisse Cucchi; Agnese Chiara Pippione; Marco L. Lolli; Barbara Giabbai; Paola Storici; Paolo Carloni; Giulia Rossetti; Federica Benvenuti; Ezia Bello; Maurizio D'Incalci; Elisa Cappuzzello; Antonio Rosato; Giannino Del Sal

The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.


Cancer Research | 2015

Abstract 5404: PAK4 allosteric modulators (PAMs) repress the Wnt/β-catenin signaling pathway and tumor growth

William Senapedis; Scott Donovan; Gali Golan; Dilara McCauley; Joel Ellis; Marsha Crochiere; Trinayan Kashyap; Boris Klebanov; Sharon Shacham; Yosef Landesman; Erkan Baloglu

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Wnt/β-catenin signaling is a ubiquitous pathway conserved throughout evolution and plays a role in embryonic and cancer development. This pathway has proven to be difficult to target therapeutically. Another intractable target is oncogenic Ras which is highly mutated in many cancer types, including pancreatic, colon and lung. An available target at the intersection of both of these genes is p21-Activated Kinase 4 (PAK4). PAK4 (1 of 6 PAK proteins) that is downstream of the Ras oncogene and a direct kinase that stabilizes transcriptional activity of β-catenin. Therefore therapeutically targeting PAK4 in cancer could be beneficial in a broad range of cancer types. PAK4 Allosteric Modulator (PAM) represents a novel and selective class of compounds that inhibit PAK4 allosterically. Methods: Flow cytometry and CellTiter AQueous One assay (MTS) were used to determine compound effects on cell cycle distribution and proliferation. CCLE, COSMIC and other databases were used for bioinformatics analysis of mutations in cancer genes. Deep sequencing and qPCR were used to analyze mRNA expression profiles. Proteomic platforms - KinomeScan, PTScan (Cell Signaling) and immunoblots were used to study phosphorylation signature of whole cell proteins and total protein steady state levels. Results: We identified a selective, orally bioavailable small molecule PAM, KPT-9274, which demonstrated anti-tumor activity in a variety of cancer cell lines (IC50 values from 0.005 - 1 μM). Bioinformatics revealed that sensitivity to KPT-9274 was directly correlated with mutations in APC or N-ras and inversely correlated with mutations in β-catenin, K-ras, or PI3K. KPT-9274 and other PAMs reduced phosphorylation and steady state levels of PAK4 protein while reducing Phospho-S675 and total β-catenin, Wnt5, Phospho- and total LRP6, Dvl2, Phospho-Akt, and Phospho-ERK. PAMs also reduced β-catenin transcriptional activity (i.e. cyclin D1, MM7, and S100A4). PAMs arrested cancer cell cycle at the G1 and G2 phases and induced apoptosis through Caspase and PARP cleavage. KPT-9274 (∼100 mg/kg BIDx5 orally) has demonstrated potent anti-tumor activity against hematological (Z-138, Molt-4, MM1S) and solid (MDA-MB-468 and Colo-205) xenograft models in mice. PAM-treated xenografts showed reduction of PAK4, β-catenin and cyclin D1 proteins. Conclusions: PAK4 represents a novel anti-cancer target at the crossroads of Ras and Wnt/β-catenin signaling. We have identified selective small molecule PAMs with anti-tumor activity both in vitro and in vivo. These allosteric modulators induce tumor cell growth arrest and apoptosis. Bioinformatics helped identify potential predictive markers in the Wnt signaling pathway while deep sequencing and proteiomics revealed possible PDn markers. Based on the in vitro and in vivo activity, KPT-9274 may be beneficial for the treatment of a wide variety of cancers and preclinical toxicology studies are ongoing. Citation Format: William T. Senapedis, Scott Donovan, Gali Golan, Dilara McCauley, Joel Ellis, Marsha Crochiere, Trinayan Kashyap, Boris Klebanov, Sharon Shacham, Yosef Landesman, Erkan Baloglu. PAK4 allosteric modulators (PAMs) repress the Wnt/β-catenin signaling pathway and tumor growth. [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 5404. doi:10.1158/1538-7445.AM2015-5404


Molecular Cancer Therapeutics | 2013

Abstract B198: Selinexor (KPT-330), a novel Selective Inhibitor of Nuclear Export (SINE) shows marked NF-kB inhibition and significant anticancer activity against Non-Small Cell Lung Cancer (NSCLC).

William Senapedis; Marsha Crochiere; Tami Rashal; Trinayan Kashyap; Boris Klebanov; Jean-Richard Saint-Martin; Ori Kalid; Sharon Shechter; Diego del Alamo; Mwanasha Hamuza; Gali Golan; Eran Shacham; Sharon Tamir; Dilara McCauley; Erkan Baloglu; Michael Kauffman; Sharon Shacham; Yosef Landesman

Selinexor (KPT-330) is a novel small molecule inhibitor of nuclear export through covalent binding to Exportin 1 (XPO1/CRM1) leading to forced nuclear retention of major tumor suppressor proteins (TSPs) such as p53, FOXO, pRB and IκB, resulting in selective death of cancer cells. Preclinical studies have shown that oral Selinexor is well tolerated, even with prolonged (4-8 month) administration. Human Phase 1 clinical studies with Selinexor in hematological and solid cancer patients are ongoing (Clinicaltrials.gov NCT01607892 and NCT01607905). In this study we evaluated the mechanisms involved in Selinexor-induced growth inhibition of NSCLC in vitro and in vivo. Methods: A panel of NSCLC cells with various genetic backgrounds were treated with Selinexor and tested in proliferation (MTT), clonogenic survival, and flow cytometric assays. The cellular localization of TSPs were evaluated by immunofluorescence (IF). Western blotting was used to study levels of cell cycle regulating proteins and quantify markers of cell survival. Functional inhibition of NF-κB was measured using a transcription factor assay kit. A SCID mouse xenograft model of NSCLC, adenocarcinoma A549 (wt p53, NF-κB IC50 of 1.19 uM), was used to evaluate Selinexor effects in vivo and tumors were evaluated by histology and immunohistochemistry (IHC). Results: Selinexor induced cell cycle arrest, inhibited tumor cell growth and clonogenic formation and induced apoptosis in lung cancer cell lines regardless of genetic background (IC50 25-700 nM). Treated normal human dermal fibroblasts entered into cell cycle arrest but resisted apoptosis induction (IC50 4.0 uM). IF of the TSPs p53, p21, IκB, E2F4, and survivin demonstrated strong nuclear localization after 4 to 24 hour treatment with Selinexor followed by apoptosis as determined by cleavage of PARP, caspases 3 and 8 by immunoblot analysis. Functional evaluation of NF-kB in A549 and H1299 revealed transcriptional repression with IC50 values similar to MTT assays. Finally, A549 xenografts treated with Selinexor (10 - 20 mg/kg QoDx3) showed marked tumor suppression when compared to vehicle and cisplatin-treated mice (growth inhibition was 78% at 20 mg/kg, p Conclusions: In NSCLC, Selinexor forces nuclear retention of TSPs, inhibits NF-κB transcriptional activity, inhibits tumor growth and induces cell death regardless of p53 status. Selinexor thus has therapeutic potential for the treatment of NSCLC. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B198. Citation Format: William Senapedis, Marsha Crochiere, Tami Rashal, Trinayan Kashyap, Boris Klebanov, Jean-Richard Saint-Martin, Ori Kalid, Sharon Shechter, Diego del Alamo, Mwanasha Hamuza, Gali Golan, Eran Shacham, Sharon Tamir, Dilara McCauley, Erkan Baloglu, Michael Kauffman, Sharon Shacham, Yosef Landesman. Selinexor (KPT-330), a novel Selective Inhibitor of Nuclear Export (SINE) shows marked NF-kB inhibition and significant anticancer activity against Non-Small Cell Lung Cancer (NSCLC). [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B198.


Molecular Cancer Therapeutics | 2013

Abstract A188: SINE resistant fibrosarcoma cells reveal changes in profile of gene expression, but continue to be sensitive to combination treatment by proteasome inhibition.

Marsha Crochiere; Trinayan Kashyap; Jean-Richard Saint-Martin; Sharon Shechter; Ori Kalid; Eran Shacham; William Senapedis; Boris Klebanov; Sharon Tamir; Diego del Alamo; Mwanasha Hamuza; Gali Golan; Erkan Baloglu; Dilara McCauley; Michael Kauffman; Sharon Shacham; Yosef Landesman

SINE (Selective Inhibitors of Nuclear Export) are novel small molecule drugs in phase I clinical trials for advanced cancers. SINEs inhibit nuclear export through covalent binding to Exportin 1 (XPO1/CRM1) leading to forced nuclear retention of major tumor suppressor proteins (TSPs) such as p53, FOXO, pRB and IkB, resulting in selective death of cancer cells. Resistant cells were created by treating the sensitive fibrosarcoma cell line HT1080 with increasing concentrations of SINE over 10 months. Gene chip analysis of parental-sensitive and drug-resistant cells treated with SINE demonstrated activation of distinct pathways that mediate either cell death or survival. In addition, SINE resistance was overcome by drug combination with proteasome inhibition. Methods: Resistant cells were generated with selection in increasing concentrations of SINE. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. FACS and immunoblots were used to measure effects on cell cycle, protein expression and cell death. RNA from nai[[Unable to Display Character: ]]ve and drug treated sensitive/resistant cells was analyzed by Affymetrix microarrays and qPCR. A drug combination study was performed to evaluate whether resistance to SINE could be overcome with proteasome inhibition. Results: Treatment of SINE-sensitive fibrosarcoma cell line HT1080 (IC50 = 13.6nM) with gradually increasing concentrations of SINE for 10 months resulted in > 100 fold decrease in sensitivity to SINE cytotoxicity (IC50 = 1.7μM). Resistant cells displayed prolonged cell cycle (∼72 vs 24 hrs) compared to parental cells. Resistant cells did not show increased MDR1 and MRP1 activity, suggesting that resistance to SINE was not mediated by induction of the multidrug resistance mechanism. Sequencing of XPO-1 from the SINE resistant cells revealed no mutations in the SINE / cargo binding pocket including the reactive Cys528. Upon exposure to SINE, resistant cells had reduced nuclear accumulation of p53, p21, FOXO1A, IkB, p27, and PP2A proteins compared with SINE-sensitive cells. SINE treatment of both sensitive and resistant cells resulted in pRB de-phosphorylation and induction of p53 and its downstream target p21. In addition, SINE treatment reduced the levels of the anti-apoptotic protein Mcl-1, and induced the apoptotic markers Caspase 3 and PARP cleavage. Microarray analysis revealed changes in cell survival and cell death pathways, which were confirmed by qPCR. In spite of the changes, resistant cells continue to show synergistic death effects induced by SINE in combination with proteasome inhibition. Conclusions: The extensive selection time (10 months) needed to achieve drug resistance suggests that generation of resistance may be difficult and that drug response may be prolonged. However such a resistance would be overcome by drug combination treatment. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A188. Citation Format: Marsha L. Crochiere, Trinayan Kashyap, Jean-Richard Saint-Martin, Sharon Shechter, Ori Kalid, Eran Shacham, William Senapedis, Boris Klebanov, Sharon Tamir, Diego del Alamo, Mwanasha Hamuza, Gali Golan, Erkan Baloglu, Dilara McCauley, Michael Kauffman, Sharon Shacham, Yosef Landesman. SINE resistant fibrosarcoma cells reveal changes in profile of gene expression, but continue to be sensitive to combination treatment by proteasome inhibition. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A188.


Cancer Research | 2013

Abstract 875: Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitors of Nuclear Export (SINE).

Yosef Landesman; Sharon Shechter; Jean-Richard Saint-Martin; Trinayan Kashyap; Marsha Crochiere; William Senapedis; Paul Ippolitti; Boris Klebanov; Sharon Tamir; Diego del Alamo; Mwanasha Hamuza; Gali Golan; Ori Kalid; Erkan Baloglu; Dilara McCauley; Michael Kauffman; Sharon Shacham

Abstract SINE are novel small molecules in human phase I clinical trials for advanced cancers. SINEs inhibit nuclear export through covalent binding to Exportin 1 (XPO1/CRM1) leading to forced nuclear retention of major tumor suppressor proteins (TSPs) such as p53, FOXO, PTEN, pRB and IKB, leading to selective death of cancer cells. Resistant cells were created by treating the sensitive fibrosarcoma cell line HT1080 with increasing concentrations of SINE over 12 months. Gene chip analysis of parental-sensitive and drug-resistant cells that were treated with SINE resulted in activation of distinct pathways that mediate mechanisms of response and drug resistance. Methods Resistant cells were generated by selection in increasing concentrations of SINE. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. FACS, qPCR and immunoblots were used to measure effects on cell cycle, gene expression and cell death. RNA from naive and drug treated sensitive/resistant cells was analyzed by Affymetrix microarrays. Results Treatment of SINE-sensitive fibrosarcoma cell line HT1080 (IC50 = 13.6nM) with gradually increasing concentrations of SINE for ∼1 year resulted with > 100 fold decrease in sensitivity to SINE cytotoxicity (IC50 = 1.7μM). Resistant cells displayed prolonged cell cycle (∼72 vs 24hrs) compared to parental cells. Resistant cells did not show increased PgP activity, suggesting that resistance to SINE was not the result of the induction of the multidrug resistance mechanism. Sequencing of CRM1 from the SINE resistant cells revealed no mutations in the SINE / cargo binding pocket including the reactive Cys528. Upon exposure to SINE, resistant cells had reduced nuclear accumulation of p53, p21, FOXO1A, IKB and p27 compared with SINE-sensitive cells. Interestingly, in SINE-sensitive cells, but not in resistant cells, SINE treatment resulted in potent pRB dephosphorylation (growth suppressive form of pRB), nuclear localization and the activation of p53 as measured by the induction of p53-downstream targets p21 and MIC1 (GDF15). In addition, SINE reduced levels of the anti-apoptotic protein Mcl-1, and induced the apoptotic markers Caspase 3 and PARP cleavage in sensitive but not in SINE resistant cells. Genes involved in cell adhesion, cytoskeleton formation, tight junctions, vesicle transport, cell cycle, and apoptosis were identified as key pathways correlating with drug response and resistance. Conclusions This study identified key pathways and genes that are likely mediating response and resistance to SINE antagonists. The extensive selection time (∼1 year) needed to achieve drug resistance suggests that generation of resistance may be difficult and that drug response may be prolonged. Citation Format: Yosef Landesman, Sharon Shechter, Jean-Richard Saint-Martin, Trinayan Kashyap, Marsha Crochiere, William Senapedis, Paul Ippolitti, Boris Klebanov, Sharon Tamir, Diego del Alamo, Mwanasha Hamuza, Gali Golan, Ori Kalid, Erkan Baloglu, Dilara McCauley, Michael Kauffman, Sharon Shacham. Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitors of Nuclear Export (SINE). [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 875. doi:10.1158/1538-7445.AM2013-875


Journal of Chemical Information and Modeling | 2014

Structure-Based Virtual Screening Approach for Discovery of Covalently Bound Ligands

Dora Warshaviak; Gali Golan; Kenneth W. Borrelli; Kai Zhu; Ori Kalid


Archive | 2016

SUBSTITUTED BENZOFURANYL AND BENZOXAZOLYL COMPOUNDS AND USES THEREOF

Erkan Baloglu; Sharon Shacham; William Senapedis; Dilara McCauley; Yosef Landesman; Gali Golan; Ori Kalid; Sharon Shechter


Archive | 2016

Substituted 2,3-Dihydrobenzofuranyl Compounds And Uses Thereof

Erkan Baloglu; Sharon Shechter; Sharon Shacham; Dilara McCauley; William Senapedis; Gali Golan; Ori Kalid


Archive | 2015

MULTICYCLIC COMPOUNDS AND METHODS OF USING SAME

Erkan Baloglu; Sharon Shacham; William Senapedis; Dilara McCauley; Yosef Landesman; Gali Golan; Ori Kalid; Sharon Shechter


Archive | 2014

BENZOFURANYLES ET BENZOXAZOLYLES SUBSTITUÉS ET LEURS UTILISATIONS

Erkan Baloglu; Sharon Shacham; William Senapedis; Dilara McCauley; Yosef Landesman; Gali Golan; Ori Kalid; Sharon Shechter

Collaboration


Dive into the Gali Golan's collaboration.

Top Co-Authors

Avatar

Erkan Baloglu

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dilara McCauley

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

William Senapedis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sharon Shechter

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yosef Landesman

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Boris Klebanov

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Trinayan Kashyap

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge