Network


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

Hotspot


Dive into the research topics where Vito J. Palombella is active.

Publication


Featured researches published by Vito J. Palombella.


Cell | 1994

The ubiquitinproteasome pathway is required for processing the NF-κB1 precursor protein and the activation of NF-κB

Vito J. Palombella; Oliver J. Rando; Alfred L. Goldberg; Tom Maniatis

Summary We demonstrate an essential role for the proteasome complex in two proteolytic processes required for activation of the transcription factor NF-κB. The p105 precursor of the p50 subunit of NF-κB is processed in vitro by an ATP-dependent process that requires proteasomes and ubiquitin conjugation. The C-terminal region of p105 is rapidly degraded, leaving the N-terminal p50 domain. p105 processing can be blocked in intact cells with inhibitors of the proteasome or in yeast with proteasome mutants. These inhibitors also block the activation of NF-κB and the rapid degradation of IκBα induced by tumor necrosis factor α. Thus, the ubiquitinproteasome pathway functions not only in the complete degradation of polypeptides, but also in the regulated processing of precursors into active proteins.


Journal of Biological Chemistry | 2002

NF-κB as a therapeutic target in multiple myeloma

Teru Hideshima; Dharminder Chauhan; Paul G. Richardson; Constantine S. Mitsiades; Nicholas Mitsiades; Toshiaki Hayashi; Nikhil C. Munshi; Lenny Dang; Alfredo Dr. Castro; Vito J. Palombella; Julian Adams; Kenneth C. Anderson

We have shown that thalidomide (Thal) and its immunomodulatory derivatives (IMiDs), proteasome inhibitor PS-341, and As2O3 act directly on multiple myeloma (MM) cells and in the bone marrow (BM) milieu to overcome drug resistance. Although Thal/IMiDs, PS-341, and As2O3 inhibit nuclear factor (NF)-κB activation, they also have multiple and varied other actions. In this study, we therefore specifically address the role of NF-κB blockade in mediating anti-MM activity. To characterize the effect of specific NF-κB blockade on MM cell growth and survival in vitro, we used an IκB kinase (IKK) inhibitor (PS-1145). Our studies demonstrate that PS-1145 and PS-341 block TNFα-induced NF-κB activation in a dose- and time-dependent fashion in MM cells through inhibition of IκBα phosphorylation and degradation of IκBα, respectively. Dexamethasone (Dex), which up-regulates IκBα protein, enhances blockade of NF-κB activation by PS-1145. Moreover, PS-1145 blocks the protective effect of IL-6 against Dex-induced apotosis. TNFα-induced intracellular adhesion molecule (ICAM)-1 expression on both RPMI8226 and MM.1S cells is also inhibited by PS-1145. Moreover, PS-1145 inhibits both IL-6 secretion from BMSCs triggered by MM cell adhesion and proliferation of MM cells adherent to BMSCs. However, in contrast to PS-341, PS-1145 only partially (20–50%) inhibits MM cell proliferation, suggesting that NF-κB blockade cannot account for all of the anti-MM activity of PS-341. Importantly, however, TNFα induces MM cell toxicity in the presence of PS-1145. These studies demonstrate that specific targeting of NF-κB can overcome the growth and survival advantage conferred both by tumor cell binding to BMSCs and cytokine secretion in the BM milieu. Furthermore, they provide the framework for clinical evaluation of novel MM therapies based upon targeting NF-κB.


Immunity | 1995

The proteasome pathway is required for cytokine-induced endothelial-leukocyte adhesion molecule expression.

Margaret Read; Andrew S. Neish; Francis W. Luscinskas; Vito J. Palombella; Tom Maniatis; Tucker Collins

Multiple cell adhesion proteins are up-regulated in vascular endothelial cells in response to TNF alpha and other inflammatory cytokines. This increase in cell adhesion gene expression is thought to require the transcription factor NF-kappa B. Here, we show that peptide aldehyde inhibitors of the proteasome, a multicatalytic protease recently shown to be required for the activation of NF-kappa B, block TNF alpha induction of the leukocyte adhesion molecules E-selectin, VCAM-1, and ICAM-1. Striking functional consequences of this inhibition were observed in analyses of leukocyte-endothelial interactions under defined flow conditions. Lymphocyte attachment to TNF alpha-treated endothelial monolayers was totally blocked, while neutrophil attachment was partially reduced but transmigration was essentially prevented.


Molecular and Cellular Biology | 2000

Nedd8 Modification of Cul-1 Activates SCFβTrCP-Dependent Ubiquitination of IκBα

Margaret Read; James E. Brownell; Tatiana B. Gladysheva; Maria Hottelet; Lana Parent; Michael B. Coggins; Jacqueline W. Pierce; Vladimir N. Podust; Rong-Shu Luo; Vincent Chau; Vito J. Palombella

ABSTRACT Regulation of NF-κB occurs through phosphorylation-dependent ubiquitination of IκBα, which is degraded by the 26S proteasome. Recent studies have shown that ubiquitination of IκBα is carried out by a ubiquitin-ligase enzyme complex called SCFβTrCP . Here we show that Nedd8 modification of the Cul-1 component of SCFβTrCP is important for function of SCFβTrCP in ubiquitination of IκBα. In cells, Nedd8-conjugated Cul-1 was complexed with two substrates of SCFβTrCP , phosphorylated IκBα and β-catenin, indicating that Nedd8–Cul-1 conjugates are part of SCFβTrCP in vivo. Although only a minute fraction of total cellular Cul-1 is modified by Nedd8, the Cul-1 associated with ectopically expressed βTrCP was highly enriched for the Nedd8-conjugated form. Moreover, optimal ubiquitination of IκBα required Nedd8 and the Nedd8-conjugating enzyme, Ubc12. The site of Nedd8 ligation to Cul-1 is essential, as SCFβTrCP containing a K720R mutant of Cul-1 only weakly supported IκBα ubiquitination compared to SCFβTrCP containing WT Cul-1, suggesting that the Nedd8 ligation of Cul-1 affects the ubiquitination activity of SCFβTrCP . These observations provide a functional link between the highly related ubiquitin and Nedd8 pathways of protein modification and show how they operate together to selectively target the signal-dependent degradation of IκBα.


Investigational New Drugs | 2000

Proteasome Inhibition: a New Strategy in Cancer Treatment

Julian Adams; Vito J. Palombella; Peter J. Elliott

The ubiquitin proteasome pathway is a highly conservedintracellular pathway for the degradation of proteins. Many of theshort-lived regulatory proteins which govern cell division, growth,activation, signaling and transcription are substrates that aretemporally degraded by the proteasome. In recent years, new andselective inhibitors of the proteasome have been employed in cellculture systems to examine the anti-tumor potential of theseagents. This review covers the chemistry of selected proteasomeinhibitors, possible mechanisms of action in cell culture and thein vivo examination of proteasome inhibitors in murine andhuman xenograft tumor models in mice. One inhibitor, PS-341, hasrecently entered Phase I clinical trials in cancer patients withadvanced disease to further test the potential of this approach.


Journal of Medicinal Chemistry | 2009

Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926).

Martin R. Tremblay; Andre Lescarbeau; Michael J. Grogan; Eddy Tan; Grace Ruiting Lin; Brian C. Austad; Lin-Chen Yu; Mark L. Behnke; Somarajan J. Nair; Margit Hagel; Kerry White; James Conley; Joseph D. Manna; Teresa M. Alvarez-Diez; Jennifer Hoyt; Caroline N. Woodward; Jens R. Sydor; Melissa Pink; John R. Macdougall; Matthew Campbell; Jill Cushing; Jeanne Ferguson; Michael Curtis; Karen McGovern; Margaret Read; Vito J. Palombella; Julian Adams; Alfredo C. Castro

Recent evidence suggests that blocking aberrant hedgehog pathway signaling may be a promising therapeutic strategy for the treatment of several types of cancer. Cyclopamine, a plant Veratrum alkaloid, is a natural product antagonist of the hedgehog pathway. In a previous report, a seven-membered D-ring semisynthetic analogue of cyclopamine, IPI-269609 (2), was shown to have greater acid stability and better aqueous solubility compared to cyclopamine. Further modifications of the A-ring system generated three series of analogues with improved potency and/or solubility. Lead compounds from each series were characterized in vitro and evaluated in vivo for biological activity and pharmacokinetic properties. These studies led to the discovery of IPI-926 (compound 28), a novel semisynthetic cyclopamine analogue with substantially improved pharmaceutical properties and potency and a favorable pharmacokinetic profile relative to cyclopamine and compound 2. As a result, complete tumor regression was observed in a Hh-dependent medulloblastoma allograft model after daily oral administration of 40 mg/kg of compound 28.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Development of 17-allylamino-17-demethoxygeldanamycin hydroquinone hydrochloride (IPI-504), an anti-cancer agent directed against Hsp90

Jens R. Sydor; Emmanuel Normant; Christine S. Pien; James R. Porter; Jie Ge; Louis Grenier; Roger H. Pak; Janid A. Ali; Marlene Dembski; Jebecka Hudak; Jon S. Patterson; Courtney Penders; Melissa Pink; Margaret Read; Jim Sang; Caroline N. Woodward; Yilong Zhang; David Grayzel; James Wright; John A. Barrett; Vito J. Palombella; Julian Adams; Jeffrey K. Tong

Heat shock protein 90 (Hsp90) is an emerging therapeutic target of interest for the treatment of cancer. Its role in protein homeostasis and the selective chaperoning of key signaling proteins in cancer survival and proliferation pathways has made it an attractive target of small molecule therapeutic intervention. 17-Allylamino-17-demethoxygeldanamycin (17-AAG), the most studied agent directed against Hsp90, suffers from poor physical-chemical properties that limit its clinical potential. Therefore, there exists a need for novel, patient-friendly Hsp90-directed agents for clinical investigation. IPI-504, the highly soluble hydroquinone hydrochloride derivative of 17-AAG, was synthesized as an Hsp90 inhibitor with favorable pharmaceutical properties. Its biochemical and biological activity was profiled in an Hsp90-binding assay, as well as in cancer-cell assays. Furthermore, the metabolic profile of IPI-504 was compared with that of 17-AAG, a geldanamycin analog currently in clinical trials. The anti-tumor activity of IPI-504 was tested as both a single agent as well as in combination with bortezomib in myeloma cell lines and in vivo xenograft models, and the retention of IPI-504 in tumor tissue was determined. In conclusion, IPI-504, a potent inhibitor of Hsp90, is efficacious in cellular and animal models of myeloma. It is synergistically efficacious with the proteasome inhibitor bortezomib and is preferentially retained in tumor tissues relative to plasma. Importantly, it was observed that IPI-504 interconverts with the known agent 17-AAG in vitro and in vivo via an oxidation-reduction equilibrium, and we demonstrate that IPI-504 is the slightly more potent inhibitor of Hsp90.


Nature | 2016

PI3Kγ is a molecular switch that controls immune suppression

Megan M. Kaneda; Karen Messer; Natacha Ralainirina; Hongying Li; Christopher J. Leem; Sara Gorjestani; Gyunghwi Woo; Abraham V. Nguyen; Camila C. Figueiredo; Philippe Foubert; Michael C. Schmid; Melissa Pink; David G. Winkler; Matthew Rausch; Vito J. Palombella; Jeffery L. Kutok; Karen McGovern; Kelly A. Frazer; Xuefeng Wu; Michael Karin; Roman Sasik; Ezra E.W. Cohen; Judith A. Varner

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPβ activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPβ activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Nature | 2016

Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells

Olivier De Henau; Matthew Rausch; David W. Winkler; Luis Felipe Campesato; Cailian Liu; Daniel Hirschhorn Cymerman; Sadna Budhu; Arnab Ghosh; Melissa Pink; Jeremy Tchaicha; Mark Douglas; Thomas T. Tibbitts; Sujata Sharma; Jennifer Proctor; Nicole Kosmider; Kerry White; Howard M. Stern; John Soglia; Julian Adams; Vito J. Palombella; Karen McGovern; Jeffery L. Kutok; Jedd D. Wolchok; Taha Merghoub

Recent clinical trials using immunotherapy have demonstrated its potential to control cancer by disinhibiting the immune system. Immune checkpoint blocking (ICB) antibodies against cytotoxic-T-lymphocyte-associated protein 4 or programmed cell death protein 1/programmed death-ligand 1 have displayed durable clinical responses in various cancers. Although these new immunotherapies have had a notable effect on cancer treatment, multiple mechanisms of immune resistance exist in tumours. Among the key mechanisms, myeloid cells have a major role in limiting effective tumour immunity. Growing evidence suggests that high infiltration of immune-suppressive myeloid cells correlates with poor prognosis and ICB resistance. These observations suggest a need for a precision medicine approach in which the design of the immunotherapeutic combination is modified on the basis of the tumour immune landscape to overcome such resistance mechanisms. Here we employ a pre-clinical mouse model system and show that resistance to ICB is directly mediated by the suppressive activity of infiltrating myeloid cells in various tumours. Furthermore, selective pharmacologic targeting of the gamma isoform of phosphoinositide 3-kinase (PI3Kγ), highly expressed in myeloid cells, restores sensitivity to ICB. We demonstrate that targeting PI3Kγ with a selective inhibitor, currently being evaluated in a phase 1 clinical trial (NCT02637531), can reshape the tumour immune microenvironment and promote cytotoxic-T-cell-mediated tumour regression without targeting cancer cells directly. Our results introduce opportunities for new combination strategies using a selective small molecule PI3Kγ inhibitor, such as IPI-549, to overcome resistance to ICB in patients with high levels of suppressive myeloid cell infiltration in tumours.


Journal of Medicinal Chemistry | 2008

Semisynthetic cyclopamine analogues as potent and orally bioavailable hedgehog pathway antagonists.

Martin R. Tremblay; Marta Nevalainen; Somarajan J. Nair; James R. Porter; Alfredo C. Castro; Mark L. Behnke; Lin-Chen Yu; Margit Hagel; Kerry White; Kerrie Faia; Louis Grenier; Matthew Campbell; Jill Cushing; Caroline N. Woodward; Jennifer Hoyt; Michael Foley; Margaret Read; Jens R. Sydor; Jeffrey K. Tong; Vito J. Palombella; Karen McGovern; Julian Adams

Herein is reported the synthesis of a novel class of hedgehog antagonists derived from cyclopamine. The acid sensitive D-ring of cyclopamine was homologated utilizing a sequence of chemoselective cyclopropanation and stereoselective acid-catalyzed rearrangement. Further modification of the A/B-ring homoallylic alcohol to the conjugated ketone led to the discovery of new cyclopamine analogues with improved pharmaceutical properties and in vitro potency (EC 50) ranging from 10 to 1000 nM.

Collaboration


Dive into the Vito J. Palombella's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julian Adams

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Janid A. Ali

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jeffery L. Kutok

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Karen McGovern

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julian Adams

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Jeffrey K. Tong

Howard Hughes Medical Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge