17 results on '"Yamini M, Ohol"'
Search Results
2. Data from Novel, Selective Inhibitors of USP7 Uncover Multiple Mechanisms of Antitumor Activity In Vitro and In Vivo
- Author
-
Paul D. Kassner, Dirk G. Brockstedt, David Wustrow, Jacob Schwarz, Christophe Colas, Jack Maung, Akinori Okano, Xinping Han, Nathan Kozon, Delia Bradford, Lisa A. Marshall, Silpa Suthram, Kyle Young, Betty Abraham, Deepa Pookot, Niket Shah, Jerick Sanchez, Steve T. Wong, Scott Jacobson, Cynthia Cho, Payal Rana, Berenger Biannic, Dennis X. Hu, Paul R. Leger, Gene Cutler, Michael T. Sun, and Yamini M. Ohol
- Abstract
The deubiquitinase USP7 regulates the levels of multiple proteins with roles in cancer progression and immune response. Thus, USP7 inhibition may decrease oncogene function, increase tumor suppressor function, and sensitize tumors to DNA-damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple TP53 wild-type cell lines, including several hematologic cancer and MYCN-amplified neuroblastoma cell lines, as well as a subset of TP53-mutant cell lines in vitro. Our work suggests that USP7 inhibitors upregulate transcription of genes normally silenced by the epigenetic repressor complex, polycomb repressive complex 2 (PRC2), and potentiate the activity of PIM and PI3K inhibitors as well as DNA-damaging agents. Furthermore, oral administration of USP7 inhibitors inhibits MM.1S (multiple myeloma; TP53 wild type) and H526 (small cell lung cancer; TP53 mutant) tumor growth in vivo. Our work confirms that USP7 is a promising, pharmacologically tractable target for the treatment of cancer.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data from Novel, Selective Inhibitors of USP7 Uncover Multiple Mechanisms of Antitumor Activity In Vitro and In Vivo
- Author
-
Paul D. Kassner, Dirk G. Brockstedt, David Wustrow, Jacob Schwarz, Christophe Colas, Jack Maung, Akinori Okano, Xinping Han, Nathan Kozon, Delia Bradford, Lisa A. Marshall, Silpa Suthram, Kyle Young, Betty Abraham, Deepa Pookot, Niket Shah, Jerick Sanchez, Steve T. Wong, Scott Jacobson, Cynthia Cho, Payal Rana, Berenger Biannic, Dennis X. Hu, Paul R. Leger, Gene Cutler, Michael T. Sun, and Yamini M. Ohol
- Abstract
Supplementary Figures S1-S6 and legends, and Tables S1, S2, S5, S6, S7.
- Published
- 2023
- Full Text
- View/download PDF
4. Novel, Selective Inhibitors of USP7 Uncover Multiple Mechanisms of Antitumor Activity In Vitro and In Vivo
- Author
-
Jacob Bradley Schwarz, Christophe Colas, Paul D. Kassner, Betty Abraham, Delia Bradford, Michael T. Sun, Lisa A. Marshall, Akinori Okano, Dennis X. Hu, Gene Cutler, Payal Rana, Paul Leger, David J. Wustrow, Cynthia Cho, Jack Maung, Yamini M. Ohol, Deepa Pookot, Nathan Kozon, Steve Wong, Scott Jacobson, Niket Shah, Berenger Biannic, Xinping Han, Silpa Suthram, Jerick Sanchez, Dirk G. Brockstedt, and Kyle Young
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Oncogene ,Chemistry ,Wild type ,Repressor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cell culture ,In vivo ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Epigenetics ,PRC2 ,PI3K/AKT/mTOR pathway - Abstract
The deubiquitinase USP7 regulates the levels of multiple proteins with roles in cancer progression and immune response. Thus, USP7 inhibition may decrease oncogene function, increase tumor suppressor function, and sensitize tumors to DNA-damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple TP53 wild-type cell lines, including several hematologic cancer and MYCN-amplified neuroblastoma cell lines, as well as a subset of TP53-mutant cell lines in vitro. Our work suggests that USP7 inhibitors upregulate transcription of genes normally silenced by the epigenetic repressor complex, polycomb repressive complex 2 (PRC2), and potentiate the activity of PIM and PI3K inhibitors as well as DNA-damaging agents. Furthermore, oral administration of USP7 inhibitors inhibits MM.1S (multiple myeloma; TP53 wild type) and H526 (small cell lung cancer; TP53 mutant) tumor growth in vivo. Our work confirms that USP7 is a promising, pharmacologically tractable target for the treatment of cancer.
- Published
- 2020
- Full Text
- View/download PDF
5. MYCN drives chemoresistance in small cell lung cancer while USP7 inhibition can restore chemosensitivity
- Author
-
Xiaoli Liu, Justin P. Norton, Deshui Jia, Renato G. Martins, Emily Eastwood, Huajia Zhang, Paul Leger, Keith D. Eaton, Eli Grunblatt, Nan Wu, Yamini M. Ohol, Rhiana Thomas, David MacPherson, A. McGarry Houghton, Joseph B. Hiatt, Ali H. Ibrahim, and Ryan Basom
- Subjects
0303 health sciences ,Chemotherapy ,biology ,medicine.medical_treatment ,Neuroendocrine Cancer ,respiratory tract diseases ,Deubiquitinating enzyme ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,Genetics ,medicine ,Cancer research ,biology.protein ,Cytotoxic T cell ,Non small cell ,neoplasms ,Research Paper ,030304 developmental biology ,Developmental Biology ,Therapeutic strategy - Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine cancer characterized by initial chemosensitivity followed by emergence of chemoresistant disease. To study roles for MYCN amplification in SCLC progression and chemoresistance, we developed a genetically engineered mouse model of MYCN-overexpressing SCLC. In treatment-naïve mice, MYCN overexpression promoted cell cycle progression, suppressed infiltration of cytotoxic T cells, and accelerated SCLC. MYCN overexpression also suppressed response to cisplatin–etoposide chemotherapy, with similar findings made upon MYCL overexpression. We extended these data to genetically perturb chemosensitive patient-derived xenograft (PDX) models of SCLC. In chemosensitive PDX models, overexpression of either MYCN or MYCL also conferred a switch to chemoresistance. To identify therapeutic strategies for MYCN-overexpressing SCLC, we performed a genome-scale CRISPR–Cas9 sgRNA screen. We identified the deubiquitinase USP7 as a MYCN-associated synthetic vulnerability. Pharmacological inhibition of USP7 resensitized chemoresistant MYCN-overexpressing PDX models to chemotherapy in vivo. Our findings show that MYCN overexpression drives SCLC chemoresistance and provide a therapeutic strategy to restore chemosensitivity.
- Published
- 2020
- Full Text
- View/download PDF
6. Direct Inhibition of Cellular Fatty Acid Synthase Impairs Replication of Respiratory Syncytial Virus and Other Respiratory Viruses.
- Author
-
Yamini M Ohol, Zhaoti Wang, George Kemble, and Gregory Duke
- Subjects
Medicine ,Science - Abstract
Fatty acid synthase (FASN) catalyzes the de novo synthesis of palmitate, a fatty acid utilized for synthesis of more complex fatty acids, plasma membrane structure, and post-translational palmitoylation of host and viral proteins. We have developed a potent inhibitor of FASN (TVB-3166) that reduces the production of respiratory syncytial virus (RSV) progeny in vitro from infected human lung epithelial cells (A549) and in vivo from mice challenged intranasally with RSV. Addition of TVB-3166 to the culture medium of RSV-infected A549 cells reduces viral spread without inducing cytopathic effects. The antiviral effect of the FASN inhibitor is a direct consequence of reducing de novo palmitate synthesis; similar doses are required for both antiviral activity and inhibition of palmitate production, and the addition of exogenous palmitate to TVB-3166-treated cells restores RSV production. TVB-3166 has minimal effect on RSV entry but significantly reduces viral RNA replication, protein levels, viral particle formation and infectivity of released viral particles. TVB-3166 substantially impacts viral replication, reducing production of infectious progeny 250-fold. In vivo, oral administration of TVB-3166 to RSV-A (Long)-infected BALB/c mice on normal chow, starting either on the day of infection or one day post-infection, reduces RSV lung titers 21-fold and 9-fold respectively. Further, TVB-3166 also inhibits the production of RSV B, human parainfluenza 3 (PIV3), and human rhinovirus 16 (HRV16) progeny from A549, HEp2 and HeLa cells respectively. Thus, inhibition of FASN and palmitate synthesis by TVB-3166 significantly reduces RSV progeny both in vitro and in vivo and has broad-spectrum activity against other respiratory viruses. FASN inhibition may alter the composition of regions of the host cell membrane where RSV assembly or replication occurs, or change the membrane composition of RSV progeny particles, decreasing their infectivity.
- Published
- 2015
- Full Text
- View/download PDF
7. Discovery of Potent, Selective, and Orally Bioavailable Inhibitors of USP7 with In Vivo Antitumor Activity
- Author
-
Deepa Pookot, Payal Rana, Paul D. Kassner, Christopher Higgs, Xinping Han, Berenger Biannic, Betty Abraham, Christophe Colas, Jacob Bradley Schwarz, Akinori Okano, Scott Jacobson, Paul Leger, Michael Sun, Jerick Sanchez, Emily Karbarz, Niket Shah, Cynthia Cho, Steve Wong, Yamini M. Ohol, Minna Bui, David J. Wustrow, Maksim Osipov, Delia Bradford, Dennis X. Hu, Jack Maung, Kyle Young, Dirk G. Brockstedt, and Grant M. Shibuya
- Subjects
Allosteric regulation ,Mutant ,Administration, Oral ,Mice, Nude ,Antineoplastic Agents ,Mice, SCID ,Crystallography, X-Ray ,01 natural sciences ,law.invention ,Ubiquitin-Specific Peptidase 7 ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Protein structure ,Succinimide ,In vivo ,law ,Mice, Inbred NOD ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Chemistry ,Wild type ,Xenograft Model Antitumor Assays ,0104 chemical sciences ,Protein Structure, Tertiary ,010404 medicinal & biomolecular chemistry ,Biochemistry ,Cell culture ,Molecular Medicine ,Suppressor - Abstract
USP7 is a promising target for cancer therapy as its inhibition is expected to decrease function of oncogenes, increase tumor suppressor function, and enhance immune function. Using a structure-based drug design strategy, a new class of reversible USP7 inhibitors has been identified that is highly potent in biochemical and cellular assays and extremely selective for USP7 over other deubiquitinases. The succinimide was identified as a key potency-driving motif, forming two strong hydrogen bonds to the allosteric pocket of USP7. Redesign of an initial benzofuran-amide scaffold yielded a simplified ether series of inhibitors, utilizing acyclic conformational control to achieve proper amine placement. Further improvements were realized upon replacing the ether-linked amines with carbon-linked morpholines, a modification motivated by free energy perturbation (FEP+) calculations. This led to the discovery of compound 41, a highly potent, selective, and orally bioavailable USP7 inhibitor. In xenograft studies, compound 41 demonstrated tumor growth inhibition in both p53 wildtype and p53 mutant cancer cell lines, demonstrating that USP7 inhibitors can suppress tumor growth through multiple different pathways.
- Published
- 2020
8. Novel, Selective Inhibitors of USP7 Uncover Multiple Mechanisms of Antitumor Activity
- Author
-
Yamini M, Ohol, Michael T, Sun, Gene, Cutler, Paul R, Leger, Dennis X, Hu, Berenger, Biannic, Payal, Rana, Cynthia, Cho, Scott, Jacobson, Steve T, Wong, Jerick, Sanchez, Niket, Shah, Deepa, Pookot, Betty, Abraham, Kyle, Young, Silpa, Suthram, Lisa A, Marshall, Delia, Bradford, Nathan, Kozon, Xinping, Han, Akinori, Okano, Jack, Maung, Christophe, Colas, Jacob, Schwarz, David, Wustrow, Dirk G, Brockstedt, and Paul D, Kassner
- Subjects
Models, Molecular ,Ubiquitin-Specific Peptidase 7 ,Mice ,Cell Line, Tumor ,Cell Culture Techniques ,Animals ,Humans ,Female - Abstract
The deubiquitinase USP7 regulates the levels of multiple proteins with roles in cancer progression and immune response. Thus, USP7 inhibition may decrease oncogene function, increase tumor suppressor function, and sensitize tumors to DNA-damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple
- Published
- 2020
9. Abstract 1646: Development of small-molecule HPK1 inhibitors to unleash tumor-specific T cell responses
- Author
-
Scott Jacobson, Jeffrey R. Jackson, Daniel Poon, Michelle Ko, Paul D. Kassner, Mikhail Zibinsky, Paul Leger, Heather Milestone, Deepika Kaveri, Anqi Ma, Thant Zaw, Lan Nguyen, Yamini M. Ohol, Tivitmahaisoon Parcharee, Chandru Ramana, Martin Brovarney, Delia Bradford, Cynthia Cho, Michael Sun, Lavanya Adusumilli, Jerick Sanchez, George E. Katibah, Omar Robles, Dan Nebalasca, Steve Wong, Blanca Gomez, Justy Gomez Guagua, Cyril Bucher, Rachel Ames, Mengshu Xu, David J. Wustrow, Anton Shakhmin, Andrew Ng, Dirk G. Brockstedt, Grant M. Shibuya, and Molly Grandcolas
- Subjects
Cancer Research ,medicine.anatomical_structure ,Oncology ,Chemistry ,T cell ,Tumor specific ,Cancer research ,medicine ,Small molecule - Abstract
Hematopoietic progenitor kinase 1 (HPK1) is an intracellular protein kinase that negatively regulates T cell signaling and proliferation. Upon T cell receptor (TCR) activation, active HPK1 phosphorylates the adaptor protein SLP76 in the TCR complex, recruiting the negative regulator 14-3-3 and targeting components of the TCR signaling complex for degradation. HPK1 thus limits the TCR signaling important for mounting an effective immune response against tumor cells. We are employing structure-guided drug design to develop potent small-molecule inhibitors of HPK1. Our compounds potently inhibit HPK1 in biochemical assays, reduce levels of phosphorylated SLP76 and concomitantly increase IL-2 production by Jurkat T cells. Importantly, our HPK1 inhibitors enhance cytokine production by human and mouse primary T cells above that observed with TCR activation alone. Treatment of mice with orally available HPK1 inhibitors results in increased activation of antigen-specific CD8+ T cells in vivo and decreased tumor growth as single agent and in combination with clinically relevant checkpoint inhibitor antibodies. Our work confirms the importance of HPK1 for T cell function and supports HPK1 as a promising next generation immuno-oncology target. Citation Format: George Katibah, Yamini Ohol, Cyril Bucher, Lavanya Adusumilli, Deepika Kaveri, Omar Robles, Michael Sun, Cynthia Cho, Heather Milestone, Rachel Ames, Scott Jacobson, Dan Nebalasca, Justy Gomez- Guagua, Jerick Sanchez, Molly Grandcolas, Steve Wong, Martin Brovarney, Chandru Ramana, Thant Zaw, Lan Nguyen, Parcharee Tivitmahaisoon, Andrew Ng, Anqi Ma, Blanca Gomez, Michelle Ko, Paul Leger, Jeffrey Jackson, Grant Shibuya, Anton Shakhmin, Delia Bradford, Mengshu Xu, Mikhail Zibinsky, Daniel Poon, David Wustrow, Paul Kassner, Dirk Brockstedt. Development of small-molecule HPK1 inhibitors to unleash tumor-specific T cell responses [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1646.
- Published
- 2021
- Full Text
- View/download PDF
10. An Mtb-Human Protein-Protein Interaction Map Reveals that Bacterial LpqN Antagonizes CBL, a Host Ubiquitin Ligase that Regulates the Balance Between Anti-Viral and Anti-Bacterial Responses
- Author
-
C Maher, M Naramura, Gwendolyn M. Jang, BH Penn, Samantha L. Bell, S Jaeger, Zoe Netter, Kristina M. Geiger, Alex Choi, Curtis Chen, Ryan D. Hernandez, Yamini M. Ohol, John Von Dollen, Tasha L. Johnson, Jeffery S. Cox, Trevor J Parry, Daniel A. Portnoy, Jeffrey R. Johnson, Michael Shales, Nevan J. Krogan, X Du, and Laurent Coscoy
- Subjects
0303 health sciences ,Innate immune system ,biology ,Host (biology) ,Mutant ,chemical and pharmacologic phenomena ,respiratory system ,biology.organism_classification ,bacterial infections and mycoses ,Molecular biology ,Ubiquitin ligase ,Cell biology ,Pathogenesis ,Mycobacterium tuberculosis ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,biology.protein ,Anti bacterial ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
SUMMARYAlthough macrophages are armed with potent anti-bacterial functions, Mycobacterium tuberculosis (Mtb) replicates inside these innate immune cells. Determinants of macrophage-intrinsic bacterial control, and the Mtb strategies to overcome them are poorly understood. To further study these processes, we used a systematic affinity tag purification mass spectrometry (AP-MS) approach to identify 187 Mtb-human protein-protein interactions (PPIs) involving 34 secreted Mtb proteins. This interaction map revealed two new factors involved in Mtb pathogenesis - the secreted Mtb protein, LpqN, and its binding partner, the human ubiquitin ligase CBL. We discovered that an lpqN Mtb mutant is attenuated in macrophages, but growth is restored when CBL is removed. Conversely, Cbl-/- macrophages are resistant to viral infection, indicating that CBL regulates cell-intrinsic polarization between anti-bacterial and anti-viral immunity. Collectively, these findings illustrate the utility of this Mtb-human PPI map as a resource for developing a deeper understanding of the intricate interactions between Mtb and its host.
- Published
- 2017
- Full Text
- View/download PDF
11. An Mtb-Human Protein-Protein Interaction Map Identifies a Switch between Host Antiviral and Antibacterial Responses
- Author
-
Zoe Netter, Jeffrey R. Johnson, Dan A. Portnoy, Laurent Coscoy, Samantha L. Bell, Kristina M. Geiger, Chen Chen, Tasha L. Johnson, Bhopal Mohapatra, Xiaotang Du, Gwendolyn M. Jang, Matthew D. Storck, Jeffery S. Cox, Bennett H. Penn, Nevan J. Krogan, Guillaume Golovkine, Alex Choi, Yamini M. Ohol, Trevor J Parry, Hamid Band, Michael Shales, Ryan D. Hernandez, Cyrus Maher, Stefanie Jäger, and John Von Dollen
- Subjects
0301 basic medicine ,Mutant ,Chlamydia trachomatis ,host-pathogen interaction ,Medical and Health Sciences ,mycobacterium ,protein-protein interaction ,Mice ,0302 clinical medicine ,Ubiquitin ,Protein Interaction Mapping ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Protein Interaction Maps ,Lymphocytes ,Proto-Oncogene Proteins c-cbl ,Aetiology ,LpqN ,Tumor ,biology ,Biological Sciences ,respiratory system ,Ubiquitin ligase ,Anti-Bacterial Agents ,Infectious Diseases ,Herpesvirus 8, Human ,Host-Pathogen Interactions ,HIV/AIDS ,Infection ,Human ,Protein Binding ,Signal Transduction ,Cbl ,Virulence Factors ,Host–pathogen interaction ,Primary Cell Culture ,chemical and pharmacologic phenomena ,macrophage ,Antiviral Agents ,Article ,Cell Line ,Microbiology ,Protein–protein interaction ,Vaccine Related ,Mycobacterium tuberculosis ,03 medical and health sciences ,Rare Diseases ,Bacterial Proteins ,Biodefense ,Cell Line, Tumor ,ubiquitin ,Tuberculosis ,Animals ,Humans ,Herpesvirus 8 ,Molecular Biology ,Innate immune system ,Prevention ,Macrophages ,HIV ,Cell Biology ,biology.organism_classification ,bacterial infections and mycoses ,Good Health and Well Being ,030104 developmental biology ,RAW 264.7 Cells ,Gene Expression Regulation ,biology.protein ,030217 neurology & neurosurgery ,Developmental Biology ,Mycobacterium - Abstract
Although macrophages are armed with potent antibacterial functions, Mycobacterium tuberculosis (Mtb) replicates inside these innate immune cells. Determinants of macrophage intrinsic bacterial control, and the Mtb strategies to overcome them, are poorly understood. To further study these processes, we used an affinity tag purification mass spectrometry (AP-MS) approach to identify 187 Mtb-human protein-protein interactions (PPIs) involving 34 secreted Mtb proteins. This interaction map revealed two factors involved in Mtb pathogenesis - the secreted Mtb protein, LpqN, and its binding partner, the human ubiquitin ligase CBL. We discovered that an lpqN Mtb mutant is attenuated in macrophages, but growth is restored when CBL is removed. Conversely, Cbl(−/−) macrophages are resistant to viral infection, indicating that CBL regulates cell-intrinsic polarization between antibacterial and antiviral immunity. Collectively, these findings illustrate the utility of this Mtb-human PPI map for developing a deeper understanding of the intricate interactions between Mtb and its host.
- Published
- 2017
12. Abstract 4441: Discovery of potent and selective inhibitors of USP7 with anti-tumor activity in vitro and in vivo
- Author
-
Nick Shah, Berenger Biannic, Andrea Kim, Scott Jacobson, Dennis X. Hu, Deepika Kaveri, Jack Maung, Gene Cutler, Betty Abraham, Jerick Sanchez, David J. Wustrow, Steve Wong, Jacob Bradley Schwarz, Akinori Okano, Paul Leger, Xinping Han, Michael Sun, Oezcan Talay, Paul D. Kassner, Delia Bradford, Cynthia Cho, Kyle Young, Deepa Pookot, Yamini M. Ohol, Nathan Kozon, Lavanya Adusumilli, Dirk G. Brockstedt, Payal Rana, and Christophe Colas
- Subjects
0301 basic medicine ,Cancer Research ,Oncogene ,Cancer ,Biology ,medicine.disease ,In vitro ,Deubiquitinating enzyme ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,Mechanism of action ,Cell culture ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,medicine.symptom - Abstract
USP7 is a deubiquitinase that regulates the levels of multiple downstream targets with roles in cancer progression and immune response. Inhibitors of USP7 may thus decrease oncogene function, increase tumor suppressor function, enhance immune function and sensitize tumor cells to DNA damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple p53-wild type cell lines, including several blood cancer and MYCN-amplified neuroblastoma cell lines, as well as a subset of p53-mutant tumor cell lines in vitro. Further, oral administration of our USP7 inhibitors inhibits MM.1S (multiple myeloma; p53-wild type) and H526 (small cell lung cancer; p53-mutant) tumor growth in vivo. Our work confirms that USP7 is a pharmacologically tractable target and future studies will aim to further understand the mechanism of action of USP7 inhibitors in p53-mutant cancers. Citation Format: Yamini M. Ohol, Michael Sun, Paul Leger, Dennis Hu, Berenger Biannic, Payal Rana, Cynthia Cho, Scott Jacobson, Steve Wong, Jerick Sanchez, Xinping Han, Kyle Young, Akinori Okano, Jack Maung, Gene Cutler, Nick Shah, Lavanya Adusumilli, Deepika Kaveri, Oezcan Talay, Deepa Pookot, Betty Abraham, Delia Bradford, Nathan Kozon, Christophe Colas, Andrea Kim, Jacob Schwarz, David Wustrow, Dirk Brockstedt, Paul Kassner. Discovery of potent and selective inhibitors of USP7 with anti-tumor activity in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4441.
- Published
- 2019
- Full Text
- View/download PDF
13. Abstract 2915: Discovery and optimization of potent and selective inhibitors of USP7 to enhance anti-tumor immunity and target tumor growth
- Author
-
Kyle Young, Jacob Bradley Schwarz, Delia Bradford, Michael Sun, Gene Cutler, Jenny McKinnell, David Chian, Cesar Meleza, Grant M. Shibuya, Silpa Suthram, Betty Abraham, Andrew Napper, Martin Brovarney, David J. Wustrow, Akinori Okano, Paul Leger, Leanne Peiser, Deepika Kaveri, Nick Shah, Xinping Han, Sherra Johnson, Deepa Pookot, Oezcan Talay, Sachie Marubayashi, Scott Jacobson, Berenger Biannic, Dennis X. Hu, Lavanya Adusumilli, Yamini M. Ohol, Jack Maung, Paul D. Kassner, Lisa A. Marshall, Angela Wadsworth, John M. Ketcham, Andrea Kim, and Payal Rana
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,Regulatory T cell ,Cell growth ,Chemistry ,FOXP3 ,medicine.anatomical_structure ,Immune system ,Oncology ,Cancer research ,medicine ,biology.protein ,PTEN ,Mdm2 ,Transcription factor - Abstract
USP7 is a deubiquitinase (DUB) that has attracted much attention recently due to its multiple roles in promoting cancer progression. By removal of ubiquitin from protein substrates, USP7 stabilizes oncogenes such as MDM2 and Myc, destabilizes and inactivates the key tumor suppressors p53 and PTEN, and imparts resistance to DNA-damaging chemotherapy by enhancing DNA repair responses. USP7 plays an important role in suppression of immune responses in the tumor microenvironment by stabilizing the transcription factor FOXP3 and thereby enhancing the suppressive function of regulatory T cells. Thus, inhibition of USP7 is an appealing therapeutic strategy because it has the potential to impact important oncology targets such as transcription factors that have been widely viewed as undruggable. We employed structure-based and other medicinal chemistry techniques to enable the design of potent and selective USP7 inhibitors. Using a high-throughput assay of DUB activity employing rhodamine-labeled ubiquitin, we optimized several series of reversible USP7 inhibitors to sub-100 pM potency and selectivity of >10,000-fold over all other DUBs. Cellular activity was demonstrated using a luciferase reporter gene assay of p53 activation, revealing compounds with EC50 values ranging down to 20 nM. To assess the role of USP7 inhibition in enhancement of immune responses, we determined relief of suppression of effector T cells in vitro. Effector T cells (CD8+) were co-cultured with regulatory T cells (CD4+ FOXP3+) and antigen-presenting cells for 4 days, after which CD8+ cell proliferation was determined by flow cytometry. Treatment with USP7 inhibitors during co-culture resulted in relief of regulatory T cell suppression of CD8+ cell proliferation. In vivo enhancement of immune responses was assessed in rodent models of inflammation and tumor growth. Direct effects on tumor cell growth and viability were explored by profiling cytotoxicity of USP7 inhibitors as single agents and in combination with chemotherapeutic agents in a broad range of cancer cell lines. In preparation for future clinical development, compounds were modified to obtain desirable in vitro and in vivo ADME and toxicity profiles. Following extensive pre-clinical optimization, we have in hand orally bioavailable compounds with high permeability, low clearance, and minimal off-target activity. Citation Format: Betty Abraham, Lavanya Adusumilli, Berenger Biannic, Delia Bradford, Martin Brovarney, David Chian, Gene Cutler, Xinping Han, Dennis Hu, Scott Jacobson, Sherra Johnson, Paul Kassner, Deepika Kaveri, John Ketcham, Andrea Kim, Paul Leger, Lisa Marshall, Sachie Marubayashi, Jack Maung, Jenny McKinnell, Cesar Meleza, Yamini Ohol, Akinori Okano, Leanne Peiser, Deepa Pookot, Payal Rana, Jacob Schwarz, Nick Shah, Grant Shibuya, Michael Sun, Silpa Suthram, Oezcan Talay, Angela Wadsworth, David Wustrow, Kyle Young, Andrew Napper. Discovery and optimization of potent and selective inhibitors of USP7 to enhance anti-tumor immunity and target tumor growth [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2915.
- Published
- 2018
- Full Text
- View/download PDF
14. Mycobacterium tuberculosis MycP1 Protease Plays a Dual Role in Regulation of ESX-1 Secretion and Virulence
- Author
-
Jeffery S. Cox, Charles S. Craik, Michael U. Shiloh, Yamini M. Ohol, David H. Goetz, and Kaman Chan
- Subjects
Cancer Research ,MICROBIO ,Virulence Factors ,medicine.medical_treatment ,Colony Count, Microbial ,Virulence ,Models, Biological ,Microbiology ,Mycobacterium tuberculosis ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,Immune system ,Bacterial Proteins ,Immunology and Microbiology(all) ,Virology ,medicine ,Animals ,Macrophage ,Secretion ,Subtilisins ,Lung ,Molecular Biology ,030304 developmental biology ,Serine protease ,Mice, Inbred BALB C ,0303 health sciences ,Protease ,biology ,030306 microbiology ,Macrophages ,Membrane Transport Proteins ,biology.organism_classification ,Survival Analysis ,3. Good health ,Chronic infection ,Liver ,biology.protein ,CELLBIO ,Parasitology ,Protein Processing, Post-Translational ,Spleen - Abstract
SummaryMycobacterium tuberculosis uses the ESX-1 secretion system to deliver virulence proteins during infection of host cells. Here we report a mechanism of posttranscriptional control of ESX-1 mediated by MycP1, a M. tuberculosis serine protease. We show that MycP1 is required for ESX-1 secretion but that, unexpectedly, genetic inactivation of MycP1 protease activity increases secretion of ESX-1 substrates. We demonstrate that EspB, an ESX-1 substrate required for secretion, is a target of MycP1 in vitro and in vivo. During macrophage infection, an inactive MycP1 protease mutant causes hyperactivation of ESX-1-stimulated innate signaling pathways. MycP1 is required for growth in mice during acute infection, while loss of its protease activity leads to attenuated virulence during chronic infection. As the key ESX-1 substrates ESAT-6 and CFP-10 are highly immunogenic, fine-tuning of their secretion by MycP1 may balance virulence and immune detection and be essential for successful maintenance of long-term M. tuberculosis infection.
- Published
- 2010
- Full Text
- View/download PDF
15. Direct Inhibition of Cellular Fatty Acid Synthase Impairs Replication of Respiratory Syncytial Virus and Other Respiratory Viruses
- Author
-
Gregory Duke, George Kemble, Zhaoti Wang, and Yamini M. Ohol
- Subjects
Rhinovirus ,viruses ,Lipoylation ,Palmitic Acid ,lcsh:Medicine ,Administration, Oral ,Gene Expression ,Respiratory Mucosa ,Respiratory Syncytial Virus Infections ,Biology ,Virus Replication ,Antiviral Agents ,Virus ,Mice ,Viral Proteins ,In vivo ,Animals ,Humans ,Enzyme Inhibitors ,lcsh:Science ,Infectivity ,Host cell membrane ,A549 cell ,chemistry.chemical_classification ,Mice, Inbred BALB C ,Multidisciplinary ,lcsh:R ,Virion ,Fatty acid ,Hep G2 Cells ,respiratory system ,Virology ,Parainfluenza Virus 3, Human ,Respiratory Syncytial Viruses ,Fatty Acid Synthase, Type I ,Fatty acid synthase ,chemistry ,Viral replication ,Host-Pathogen Interactions ,biology.protein ,lcsh:Q ,Protein Processing, Post-Translational ,HeLa Cells ,Research Article - Abstract
Fatty acid synthase (FASN) catalyzes the de novo synthesis of palmitate, a fatty acid utilized for synthesis of more complex fatty acids, plasma membrane structure, and post-translational palmitoylation of host and viral proteins. We have developed a potent inhibitor of FASN (TVB-3166) that reduces the production of respiratory syncytial virus (RSV) progeny in vitro from infected human lung epithelial cells (A549) and in vivo from mice challenged intranasally with RSV. Addition of TVB-3166 to the culture medium of RSV-infected A549 cells reduces viral spread without inducing cytopathic effects. The antiviral effect of the FASN inhibitor is a direct consequence of reducing de novo palmitate synthesis; similar doses are required for both antiviral activity and inhibition of palmitate production, and the addition of exogenous palmitate to TVB-3166-treated cells restores RSV production. TVB-3166 has minimal effect on RSV entry but significantly reduces viral RNA replication, protein levels, viral particle formation and infectivity of released viral particles. TVB-3166 substantially impacts viral replication, reducing production of infectious progeny 250-fold. In vivo, oral administration of TVB-3166 to RSV-A (Long)-infected BALB/c mice on normal chow, starting either on the day of infection or one day post-infection, reduces RSV lung titers 21-fold and 9-fold respectively. Further, TVB-3166 also inhibits the production of RSV B, human parainfluenza 3 (PIV3), and human rhinovirus 16 (HRV16) progeny from A549, HEp2 and HeLa cells respectively. Thus, inhibition of FASN and palmitate synthesis by TVB-3166 significantly reduces RSV progeny both in vitro and in vivo and has broad-spectrum activity against other respiratory viruses. FASN inhibition may alter the composition of regions of the host cell membrane where RSV assembly or replication occurs, or change the membrane composition of RSV progeny particles, decreasing their infectivity.
- Published
- 2015
16. Patellin1, a Novel Sec14-Like Protein, Localizes to the Cell Plate and Binds Phosphoinositides
- Author
-
T. Kaye Peterman, Yamini M. Ohol, Lisa J. McReynolds, and Elizabeth J. Luna
- Subjects
Cell division ,Physiology ,Molecular Sequence Data ,Arabidopsis ,Plant Science ,Plasma protein binding ,Biology ,Phosphatidylinositols ,Plant Roots ,chemistry.chemical_compound ,symbols.namesake ,Gene Expression Regulation, Plant ,Tobacco ,Genetics ,Amino Acid Sequence ,Phosphatidylinositol ,Phospholipid Transfer Proteins ,Sequence Homology, Amino Acid ,Arabidopsis Proteins ,Cell plate ,Golgi apparatus ,Cell biology ,Microscopy, Fluorescence ,chemistry ,Cytoplasm ,Multigene Family ,symbols ,Sequence Alignment ,Biogenesis ,Cytokinesis ,Protein Binding ,Research Article - Abstract
Membrane trafficking is central to construction of the cell plate during plant cytokinesis. Consequently, a detailed understanding of the process depends on the characterization of molecules that function in the formation, transport, targeting, and fusion of membrane vesicles to the developing plate, as well as those that participate in its consolidation and maturation into a fully functional partition. Here we report the initial biochemical and functional characterization of patellin1 (PATL1), a novel cell-plate-associated protein that is related in sequence to proteins involved in membrane trafficking in other eukaryotes. Analysis of the Arabidopsis genome indicated that PATL1 is one of a small family of Arabidopsis proteins, characterized by a variable N-terminal domain followed by two domains found in other membrane-trafficking proteins (Sec14 and Golgi dynamics domains). Results from immunolocalization and biochemical fractionation studies suggested that PATL1 is recruited from the cytoplasm to the expanding and maturing cell plate. In vesicle-binding assays, PATL1 bound to specific phosphoinositides, important regulators of membrane trafficking, with a preference for phosphatidylinositol(5)P, phosphatidylinositol(4,5)P2, and phosphatidylinositol(3)P. Taken together, these findings suggest a role for PATL1 in membranetrafficking events associated with cell-plate expansion or maturation and point to the involvement of phosphoinositides in cell-plate biogenesis. Plant cells partition their cytoplasm during cytokinesis by building a new cell wall from the insideout between the two sets of daughter chromosomes (Staehelin and Hepler, 1996; Heese et al., 1998; Smith
- Published
- 2004
- Full Text
- View/download PDF
17. Role for lysosomal enzyme β-hexosaminidase in the control of mycobacteria infection
- Author
-
Ping Wu, Eric J. Brown, J. Hiroshi Morisaki, Ingrid Chou Koo, Yamini M. Ohol, and Jeffery S. Cox
- Subjects
Mice, Transgenic ,Microbiology ,Cell Line ,Mice ,Immunity ,RNA interference ,Lysosome ,medicine ,Macrophage ,Animals ,Humans ,Secretion ,Microscopy, Phase-Contrast ,Mycobacterium marinum ,Mycobacterium Infections ,Multidisciplinary ,biology ,Macrophages ,Biological Sciences ,biology.organism_classification ,beta-N-Acetylhexosaminidases ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cell culture ,Drosophila ,RNA Interference ,Lysosomes ,Dimerization ,Intracellular - Abstract
The pathogenic mycobacteria that cause tuberculosis (TB) and TB-like diseases in humans and animals elude sterilizing immunity by residing within an intracellular niche in host macrophages, where they are protected from microbicidal attack. Recent studies have emphasized microbial mechanisms for evasion of host defense; less is known about mycobactericidal mechanisms that remain intact during initial infection. To better understand macrophage mechanisms for restricting mycobacteria growth, we examined Mycobacterium marinum infection of Drosophila S2 cells. Among ≈1,000 host genes examined by RNAi depletion, the lysosomal enzyme β-hexosaminidase was identified as an important factor in the control of mycobacterial infection. The importance of β-hexosaminidase for restricting mycobacterial growth during mammalian infections was confirmed in macrophages from β-hexosaminidase knockout mice. β-Hexosaminidase was characterized as a peptidoglycan hydrolase that surprisingly exerts its mycobactericidal effect at the macrophage plasma membrane during mycobacteria-induced secretion of lysosomes. Thus, secretion of lysosomal enzymes is a mycobactericidal mechanism that may have a more general role in host defense.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.