Network


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

Hotspot


Dive into the research topics where Charles Voliva is active.

Publication


Featured researches published by Charles Voliva.


ACS Medicinal Chemistry Letters | 2011

Identification of NVP-BKM120 as a Potent, Selective, Orally Bioavailable Class I PI3 Kinase Inhibitor for Treating Cancer

Matthew Burger; Sabina Pecchi; Allan S. Wagman; Zhi-Jie Ni; Mark Knapp; Thomas Hendrickson; Gordana Atallah; Keith B. Pfister; Yanchen Zhang; Sarah Bartulis; Kelly Frazier; Simon Ng; Aaron Smith; Joelle Verhagen; Joshua Haznedar; Kay Huh; Ed Iwanowicz; Xiaohua Xin; Daniel Menezes; Hanne Merritt; Isabelle Lee; Marion Wiesmann; Susan Kaufman; Kenneth Crawford; Michael Chin; Dirksen E. Bussiere; Kevin Shoemaker; Isabel Zaror; Sauveur-Michel Maira; Charles Voliva

Phosphoinositide-3-kinases (PI3Ks) are important oncology targets due to the deregulation of this signaling pathway in a wide variety of human cancers. Herein we describe the structure guided optimization of a series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines where the pharmacokinetic properties were improved by modulating the electronics of the 6-position heterocycle, and the overall druglike properties were fine-tuned further by modification of the 4-position substituent. The resulting 2,4-bismorpholino 6-heterocyclic pyrimidines are potent class I PI3K inhibitors showing mechanism modulation in PI3K dependent cell lines and in vivo efficacy in tumor xenograft models with PI3K pathway deregulation (A2780 ovarian and U87MG glioma). These efforts culminated in the discovery of 15 (NVP-BKM120), currently in Phase II clinical trials for the treatment of cancer.


Cancer Research | 2010

Abstract 4498: Biological characterization of NVP-BKM120, a novel inhibitor of phosphoinosotide 3-kinase in Phase I/II clinical trials

Charles Voliva; Sabina Pecchi; Matthew Burger; Tobi Nagel; Christian Schnell; Christine Fritsch; Saskia M. Brachmann; Daniel Menezes; Mark Knapp; Kevin Shoemaker; Marion Wiesmann; Kay Huh; Isabel Zaror; Marion Dorsch; William R. Sellers; Carlos Garcia-Echeverria; Michel Maira

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The PI3K/Akt/mTor signaling pathway plays an important role in controlling cell growth, proliferation and survival. Through various mechanisms, the pathway is frequently dysregulated in human cancers, suggesting the use of PI3K inhibitors as novel targeted anticancer therapeutic agents. To this end, substantial drug discovery efforts have been devoted both in pharmaceutical companies and in academia to identify and develop therapeutic agents able to specifically down regulate PI3K or other components of this pathway in tumors cells. Following the discovery of NVP-BEZ235, our first dual pan-PI3K/mTOR clinical compound, we sought to identify additional PI3K inhibitors from different chemical classes with more stringent selectivity profiles. The key to achieve these objectives was to pursue a structure-based design approach coupled with intensive pharmacological evaluation of selected compounds during the medicinal chemistry optimization process. Here we report on the biological characterization of the pan-PI3K pyrimidine-derived inhibitor NVP-BKM120. This compound inhibits all four Class I PI3K isoforms (IC50 values in the 35 to 248 nM range) with at least 50-fold selectivity (compared to p110α) towards protein kinases. The compound is also active against the most common somatic PI3Kα mutations (H1047R, E542K and E545K). NVP-BKM120 does not significantly inhibit the related Class III (Vps34) and Class IV (mTOR, DNA-PK) PI3K kinases. Consistent with its mechanism of action, NVP-BKM120 decreases the cellular levels of p-Akt in mechanistic and relevant tumor cell lines (e.g., IC50 for S473P-Akt in Rat1-p110α cells of 93 nM). This biological activity correlates with inhibition of various Akt downstream signaling pathway components, and with its anti-proliferative activity. Thus, the compound demonstrates significant, concentration dependent cell growth inhibition and induction of apoptosis in a variety of tumor cancer cells, particularly for those harboring p110α mutants and/or over-expressing erbB2. In addition, NVP-BKM120 demonstrates significant, dose dependent in vivo pharmacodynamic activity as measured by inhibition of p-Akt in relevant xenograft models. The pharmacological, biological and preclinical safety profile of NVP-BKM120 supports its clinical development and the compound is currently undergoing Phase 1/II clinical trials in cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4498.


ACS Medicinal Chemistry Letters | 2011

Synthesis and in Vitro and in Vivo Evaluation of Phosphoinositide-3-kinase Inhibitors.

Matthew Burger; Mark Knapp; Allan S. Wagman; Zhi-Jie Ni; Thomas Hendrickson; Gordana Atallah; Yanchen Zhang; Kelly Frazier; Joelle Verhagen; Keith B. Pfister; Simon Ng; Aaron Smith; Sarah Bartulis; Hanne Merrit; Marion Weismann; Xiaohua Xin; Joshua Haznedar; Charles Voliva; Ed Iwanowicz; Sabina Pecchi

Phospoinositide-3-kinases (PI3K) are important oncology targets due to the deregulation of this signaling pathway in a wide variety of human cancers. A series of 2-morpholino, 4-substituted, 6-(3-hydroxyphenyl) pyrimidines have been reported as potent inhibitors of PI3Ks. Herein, we describe the structure-guided optimization of these pyrimidines with a focus on replacing the phenol moiety, while maintaining potent target inhibition and improving in vivo properties. A series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines, which potently inhibit PI3K, were discovered. Within this series a compound, 17, was identified with suitable pharmacokinetic (PK) properties, which allowed for the establishment of a PI3K PK/pharmacodynamic-efficacy relationship as determined by in vivo inhibition of AKT(Ser473) phosphorylation and tumor growth inhibition in a mouse A2780 tumor xenograft model.


Bioorganic & Medicinal Chemistry Letters | 2010

Identification and structure-activity relationship of 2-morpholino 6-(3-hydroxyphenyl) pyrimidines, a class of potent and selective PI3 kinase inhibitors.

Sabina Pecchi; Paul A. Renhowe; Clarke Taylor; Susan Kaufman; Hanne Merritt; Marion Wiesmann; Kevin Shoemaker; Mark Knapp; Elizabeth Ornelas; Thomas Hendrickson; Wendy J. Fantl; Charles Voliva

PI3 Kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration, and differentiation. The PI3 kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations, and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe a novel series of PI3K inhibitors sharing a pyrimidine core and showing significant potency against class I PI3 kinases in the biochemical assay and in cells. The discovery, synthesis and SAR of this chemotype are described.


ACS Medicinal Chemistry Letters | 2015

Optimization of a Dibenzodiazepine Hit to a Potent and Selective Allosteric PAK1 Inhibitor.

Alexei Karpov; Payman Amiri; Cornelia Bellamacina; Marie-Helene Bellance; Werner Breitenstein; Dylan Daniel; Regis Denay; Doriano Fabbro; César Fernández; Inga Galuba; Stephanie Guerro-Lagasse; Sascha Gutmann; Linda Hinh; Wolfgang Jahnke; Julia Klopp; Albert Lai; Mika Lindvall; Sylvia Ma; Henrik Möbitz; Sabina Pecchi; Gabriele Rummel; Kevin Shoemaker; Joerg Trappe; Charles Voliva; Sandra W. Cowan-Jacob; Andreas Marzinzik

The discovery of inhibitors targeting novel allosteric kinase sites is very challenging. Such compounds, however, once identified could offer exquisite levels of selectivity across the kinome. Herein we report our structure-based optimization strategy of a dibenzodiazepine hit 1, discovered in a fragment-based screen, yielding highly potent and selective inhibitors of PAK1 such as 2 and 3. Compound 2 was cocrystallized with PAK1 to confirm binding to an allosteric site and to reveal novel key interactions. Compound 3 modulated PAK1 at the cellular level and due to its selectivity enabled valuable research to interrogate biological functions of the PAK1 kinase.


Bioorganic & Medicinal Chemistry Letters | 2013

Structure guided optimization of a fragment hit to imidazopyridine inhibitors of PI3K.

Sabina Pecchi; Zhi-Jie Ni; Wooseok Han; Aaron Smith; Jiong Lan; Matthew Burger; Hanne Merritt; Marion Wiesmann; John Chan; Susan Kaufman; Mark Knapp; Johanna Janssen; Kay Huh; Charles Voliva

PI3 kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration and differentiation. The PI3 Kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe how the potency of a benzothiazole fragment hit was quickly improved based on structural information and how this early chemotype was further optimized through scaffold hopping. This effort led to the identification of a series of 2-acetamido-5-heteroaryl imidazopyridines showing potent in vitro activity against all class I PI3Ks and attractive pharmacokinetic properties.


Cancer Research | 2010

Abstract 4472: NVP-BKM120, a pan class I PI3K inhibitor impairs microvascular permeability and tumor growth as detected by DCE-MRI and IFP measurements via radio-telemetry: Comparison with NVP-BEZ235

Christian Schnell; Samuel Arnal; Mike Becquet; Peter R. Allegrini; Charles Voliva; Robert Cozens; Carlos Garcia-Echeverria; Sauveur Michel Maira

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Tumor blood vessels are distinctly abnormal in structure and function. When compared to normal vessels, they are enlarged and tortuous and leaky, poorly covered by pericytes and with a defective basement membrane. VEGF is a prominent cytokine responsible for the hyperpermeable state of tumor microvasculature to plasma macromolecules. The phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway functions downstream of VEGF and can activate the production of the free radical gas nitric oxide (NO) through the enzyme endothelial NO synthase (eNOS), a key player for the in vivo biological activity of VEGF. In the present study, we investigated the effect of NVP-BKM120, a novel, synthetic low molecular mass compound, which potently inhibits class I PI3K activity, on vascular leakage in tumors and its impact on tumor progression in an orthotopic breast cancer model (BN472) in syngeneic rats. Effects on the vasculature were investigated in normal and tumor tissues with a variety of technologies: dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), tumor interstitial fluid pressure (IFP) measurements in conscious animals via radio-telemetry and a modified Miles assay, representing non invasive, minimally invasive and invasive methods, respectively. Tumor histology and western blots were used to confirm pathway inhibition. For comparison, the effect of NVP-BEZ235, a potent pan class I PI3K and mTOR activity inhibitor, was assessed head-to-head in the same experiments. BKM120 induced significant decreases in VEGF-induced vascular leakage in normal and tumor tissues. These effects were similar to those obtained following BEZ235 treatment. Western blot of total ear lysates from treated mice clearly demonstrated pathway inhibition. The inhibition of vessel permeability observed with BEZ235 and BKM120 correlated with their respective anti-tumor activities, IFP change and Ktrans (vascular permeability transfer constant measured by DCE-MRI) decrease. In conclusion, our data demonstrate that in BN472 tumors, BKM120 significantly decreases tumor microvascular leakage similar to BEZ235 at equipotent antitumor dose. Changes in Ktrans may be a useful independent clinical surrogate biomarker for NVP-BKM120 efficacy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4472.


Cancer Research | 2014

Abstract LB-121: Dissecting MAPK pathway in BRAFmut melanoma: Intricacies of ERK1 and ERK2

Tatiana Zavorotinskaya; Upasana Mehra; Yumin Dai; Michel Faure; Ken Crawford; Karen Yu; Jan Marie Cheng; Xiaolei Ma; Jan Xuan; Kelly Yan; Mohammad Hekmat-Nejad; Hanne Merritt; Darrin Stuart; Charles Voliva

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The MAPK signaling cascade, comprised of the RAS GTPases, the RAF, MEK1/2 and ERK1/2 kinases is frequently deregulated in cancer. ERK1 and ERK2 transmit signals generated by mutant BRAF, Ras or by activated receptor tyrosine kinases to a wide range of nuclear and cytoplasmic substrates, resulting in signal amplification, cell growth, migration and survival. ERK1 and ERK2 have been considered as redundant because of their high homology, large number of overlapping substrates, and ability to substitute for each other in genetically engineered mouse models. Nevertheless, several investigators have identified non-redundant roles for ERK isoforms in oncogenesis; for instance, ERK2, but not ERK1, appears to be responsible for RASmut induced epithelial-to-mesenchymal transformation. Besides, each of the ERK isoforms employs spatially distinct substrate docking domains, DEF (docking site for ERK FXFP) and D (docking domain), to signal to different subsets of substrates and differentially transmit signals downstream. We set out to determine the roles of ERK isoforms as well as DEF- and D-domain dependent signaling in the survival of melanoma tumor cells expressing activating BRAF mutations which are highly sensitive to pharmacological inhibitors of RAF, MEK1/2 and ERK1/2. We designed ERK1 and ERK2 mutants resistant to ATP-competitive ERK1/2 inhibitors and employed auto-activating, MEK-independent, ERK1 and ERK2 mutants to ask if BRAFmut melanoma survival is dependent on either or both ERK isoforms. In addition, we used RNAi and zinc-finger nucleases’ to knockdown or delete each of ERK isoforms. These experimental approaches consistently demonstrated that ERK2, but not ERK1, was the sole driver of cell survival in multiple BRAFmut melanoma cell lines. Moreover, genome-wide gene expression analysis indicated that ERK2, but not ERK1, was largely responsible for transcriptional effects imposed by pharmacological RAF, MEK1/2 or ERK1/2 inhibitors. Thus, in BRAFmut melanoma, functions of ERK1 and 2 are not redundant, and ERK1 cannot substitute for a disabled ERK2. Next, we introduced DEF- and D-substrate docking domain mutations into an ERK inhibitor resistant ERK2 to investigate whether signaling through either domain is sufficient to support melanoma survival. We observed that signaling through D- or DEF- domains of ERK2 had differential effects on gene expression and substrate phosphorylation. Consequently, we have found that a subset of melanoma cell lines was sensitive to elimination of DEF- docking domain interactions, whereas another subset of cell lines tolerated mutations in the DEF-site. Interactions and signaling through ERK D-docking site were dispensable for survival of all melanoma cell lines tested. These data suggest potential novel approaches to target oncogenic MAPK pathway. Citation Format: Tatiana Zavorotinskaya, Upasana Mehra, Yumin Dai, Michel Faure, Ken Crawford, Karen Yu, Jan Marie Cheng, Xiaolei Ma, Jan Xuan, Kelly Yan, Mohammad Hekmat-Nejad, Hanne Merritt, Darrin Stuart, Charles Voliva. Dissecting MAPK pathway in BRAFmut melanoma: Intricacies of ERK1 and ERK2. [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 LB-121. doi:10.1158/1538-7445.AM2014-LB-121


Cancer Research | 2010

Abstract 4497: NVP-BKM120, a novel inhibitor of phosphoinosotide 3-kinase in Phase I/II clinical trials, shows significant antitumor activity in xenograft and primary tumor models

Michel Maira; Daniel Menezes; Sabina Pecchi; Kevin Shoemaker; Matthew Burger; Christian Schnell; Christine Fritsch; Saskia M. Brachmann; Tobi Nagel; William R. Sellers; Carlos Garcia-Echeverria; Marion Wiesmann; Charles Voliva

A substantial number of epidemiological and experimental studies support an important role for PI3K in the biology of human cancer. The activation of PI3K, and its downstream effectors, has been clearly validated as an essential step for the initiation and maintenance of the tumorigenic phenotype. Parallel to the clinical development of our dual pan-PI3K/mTOR modulators (e.g., NVP-BEZ235), we continued our drug discovery activities to identify PI3K inhibitors with distinct biological and pharmacological profiles. Following a structure-based design strategy, we have identified a new clinical candidate, NVP-BKM120. This 2,6-dimorpholino pyrimidine derivative is a potent pan-PI3K (e.g., IC 50 = 35 nM, p110α) that does not significantly inhibit other protein or lipid kinases (e.g., IC 50 = 4.6 μM, mTOR). The compound exhibits potent antiproliferative activity against a broad panel of tumour cell lines by specifically blocking the biological function of PI3K signaling components (e.g. IC 50 = 93 nM S473P-Akt in Rat1-p110α cells). The activity of this pan-PI3K inhibitor in cellular settings translates well in in vivo models of human cancer. NVP-BKM120 shows good oral bioavailability in preclinical species and demonstrated significant antitumor activity (growth inhibition or regression) at tolerated doses in xenografts of diverse cancer lineage in mice and rat models. Consistent with its mechanism of action, transient increases in plasma insulin and glucose levels are observed in these studies. Analyses of tumor tissues after acute dosing or at the end of efficacy studies showed a good correlation between compound exposure, PI3K pathway blockade (reduction in P-Akt levels) and antitumor activity. Dose dependent tumor growth delay or regression is also observed in primary tumor models, and, as demonstrated previously with our dual pan-PI3K/mTOR inhibitor NVP-BEZ235, in vivo synergistic activity is observed when NVP-BKM120 is combined with a MEK inhibitor in K-Ras human cancer models. NVP-BKM120 is currently undergoing Phase I/II human clinical trials for the treatment of solid tumors and hematological malignancies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4497.


Archive | 2007

Pyrimidine derivatives used as PI-3-kinase inhibitors

Matthew Burger; Zhi-Jie Ni; Sabina Pecchi; Gordana Atallah; Sarah Bartulis; Kelly Frazier; Aaron Smith; Joelle Verhagen; Yanchen Zhang; Allan S. Wagman; Simon Ng; Keith B. Pfister; Daniel J. Poon; Alicia Louie; Teresa E. Pick; Paul A. Barsanti; Edwin Iwanowicz; Wendy J. Fantl; Thomas Hendrickson; Mark Knapp; Hanne Meritt; Charles Voliva; Marion Wiesmann; Xiahua Xin

Collaboration


Dive into the Charles Voliva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge