Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Natl Cancer Inst ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710487

ABSTRACT

BACKGROUND: Camonsertib is a selective oral inhibitor of ataxia telangiectasia and Rad3-related (ATR) kinase with demonstrated efficacy in tumors with DNA damage response gene deficiencies. On-target anemia is the main drug-related toxicity typically manifesting after the period of dose-limiting toxicity evaluation. Thus dose/schedule optimization requires extended follow-up to assess prolonged treatment effects. METHODS: Long-term safety/tolerability and antitumor efficacy of three camonsertib monotherapy dose levels/schedules were assessed in the TRESR study dose-optimization phase: 160 mg once daily (QD) 3 days on/4 off (160 3/4; the preliminary recommended phase II dose [RP2D]) and two step-down groups of 120 mg QD 3/4 (120 3/4) and 160 mg QD 3/4, 2 weeks on/1 off (160 3/4, 2/1w). Safety endpoints included incidence of treatment-related adverse events (TRAEs), dose modifications, and transfusions. Efficacy endpoints included overall response rate, clinical benefit rate, progression-free survival, and circulating-tumor-DNA (ctDNA)-based molecular response rate. RESULTS: The analysis included 119 patients: 160 3/4 (n = 67), 120 3/4 (n = 25), and 160 3/4, 2/1w (n = 27) treated up to 117.1 weeks as of the data cutoff. The risk of developing grade 3 anemia was significantly lower in the 160 3/4, 2/1w group compared with the preliminary RP2D group (HR = 0.23, 2-sided P = .02), translating to reduced transfusion and dose reduction requirements. The intermittent weekly schedule did not compromise antitumor activity. CONCLUSION: The 160 3/4, 2/1w dose was established as an optimized regimen for future camonsertib monotherapy studies offering significantly reduced anemia incidence without any compromise to efficacy.

2.
Nat Med ; 29(6): 1400-1411, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37277454

ABSTRACT

Predictive biomarkers of response are essential to effectively guide targeted cancer treatment. Ataxia telangiectasia and Rad3-related kinase inhibitors (ATRi) have been shown to be synthetic lethal with loss of function (LOF) of ataxia telangiectasia-mutated (ATM) kinase, and preclinical studies have identified ATRi-sensitizing alterations in other DNA damage response (DDR) genes. Here we report the results from module 1 of an ongoing phase 1 trial of the ATRi camonsertib (RP-3500) in 120 patients with advanced solid tumors harboring LOF alterations in DDR genes, predicted by chemogenomic CRISPR screens to sensitize tumors to ATRi. Primary objectives were to determine safety and propose a recommended phase 2 dose (RP2D). Secondary objectives were to assess preliminary anti-tumor activity, to characterize camonsertib pharmacokinetics and relationship with pharmacodynamic biomarkers and to evaluate methods for detecting ATRi-sensitizing biomarkers. Camonsertib was well tolerated; anemia was the most common drug-related toxicity (32% grade 3). Preliminary RP2D was 160 mg weekly on days 1-3. Overall clinical response, clinical benefit and molecular response rates across tumor and molecular subtypes in patients who received biologically effective doses of camonsertib (>100 mg d-1) were 13% (13/99), 43% (43/99) and 43% (27/63), respectively. Clinical benefit was highest in ovarian cancer, in tumors with biallelic LOF alterations and in patients with molecular responses. ClinicalTrials.gov registration: NCT04497116 .


Subject(s)
Ataxia Telangiectasia , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Protein Kinase Inhibitors/pharmacokinetics , DNA Damage , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
3.
NPJ Syst Biol Appl ; 3: 16034, 2017.
Article in English | MEDLINE | ID: mdl-28725482

ABSTRACT

The ErbB family of receptor tyrosine kinases comprises four members: epidermal growth factor receptor (EGFR/ErbB1), human EGFR 2 (HER2/ErbB2), ErbB3/HER3, and ErbB4/HER4. The first two members of this family, EGFR and HER2, have been implicated in tumorigenesis and cancer progression for several decades, and numerous drugs have now been approved that target these two proteins. Less attention, however, has been paid to the role of this family in mediating cancer cell survival and drug tolerance. To better understand the complex signal transduction network triggered by the ErbB receptor family, we built a computational model that quantitatively captures the dynamics of ErbB signaling. Sensitivity analysis identified ErbB3 as the most critical activator of phosphoinositide 3-kinase (PI3K) and Akt signaling, a key pro-survival pathway in cancer cells. Based on this insight, we designed a fully human monoclonal antibody, seribantumab (MM-121), that binds to ErbB3 and blocks signaling induced by the extracellular growth factors heregulin (HRG) and betacellulin (BTC). In this article, we present some of the key preclinical simulations and experimental data that formed the scientific foundation for three Phase 2 clinical trials in metastatic cancer. These trials were designed to determine if patients with advanced malignancies would derive benefit from the addition of seribantumab to standard-of-care drugs in platinum-resistant/refractory ovarian cancer, hormone receptor-positive HER2-negative breast cancer, and EGFR wild-type non-small cell lung cancer (NSCLC). From preclinical studies we learned that basal levels of ErbB3 phosphorylation correlate with response to seribantumab monotherapy in mouse xenograft models. As ErbB3 is rapidly dephosphorylated and hence difficult to measure clinically, we used the computational model to identify a set of five surrogate biomarkers that most directly affect the levels of p-ErbB3: HRG, BTC, EGFR, HER2, and ErbB3. Preclinically, the combined information from these five markers was sufficient to accurately predict which xenograft models would respond to seribantumab, and the single-most accurate predictor was HRG. When tested clinically in ovarian, breast and lung cancer, HRG mRNA expression was found to be both potentially prognostic of insensitivity to standard therapy and potentially predictive of benefit from the addition of seribantumab to standard of care therapy in all three indications. In addition, it was found that seribantumab was most active in cancers with low levels of HER2, consistent with preclinical predictions. Overall, our clinical studies and studies of others suggest that HRG expression defines a drug-tolerant cancer cell phenotype that persists in most solid tumor indications and may contribute to rapid clinical progression. To our knowledge, this is the first example of a drug designed and clinically tested using the principles of Systems Biology.

4.
PLoS One ; 7(9): e46485, 2012.
Article in English | MEDLINE | ID: mdl-23029530

ABSTRACT

Expression of the hominoid-specific TBC1D3 oncoprotein enhances growth factor receptor signaling and subsequently promotes cellular proliferation and survival. Here we report that TBC1D3 is degraded in response to growth factor signaling, suggesting that TBC1D3 expression is regulated by a growth factor-driven negative feedback loop. To gain a better understanding of how TBC1D3 is regulated, we studied the effects of growth factor receptor signaling on TBC1D3 post-translational processing and turnover. Using a yeast two-hybrid screen, we identified CUL7, the scaffolding subunit of the CUL7 E3 ligase complex, as a TBC1D3-interacting protein. We show that CUL7 E3 ligase ubiquitinates TBC1D3 in response to serum stimulation. Moreover, TBC1D3 recruits F-box 8 (Fbw8), the substrate recognition domain of CUL7 E3 ligase, in pull-down experiments and in an in vitro assay. Importantly, alkaline phosphatase treatment of TBC1D3 suppresses its ability to recruit Fbw8, indicating that TBC1D3 phosphorylation is critical for its ubiquitination and degradation. We conclude that serum- and growth factor-stimulated TBC1D3 ubiquitination and degradation are regulated by its interaction with CUL7-Fbw8.


Subject(s)
Cullin Proteins/metabolism , F-Box Proteins/metabolism , GTPase-Activating Proteins/metabolism , Protein Processing, Post-Translational , Proteolysis , Proto-Oncogene Proteins/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/physiology , Leupeptins/pharmacology , Phosphorylation , Proteasome Inhibitors/pharmacology , Protein Binding , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
PLoS One ; 7(2): e31225, 2012.
Article in English | MEDLINE | ID: mdl-22348058

ABSTRACT

Insulin/IGF-1 signaling plays a pivotal role in the regulation of cellular homeostasis through its control of glucose metabolism as well as due to its effects on cell proliferation. Aberrant regulation of insulin signaling has been repeatedly implicated in uncontrolled cell growth and malignant transformations. TBC1D3 is a hominoid specific gene previously identified as an oncogene in breast and prostate cancers. Our efforts to identify the molecular mechanisms of TBC1D3-induced oncogenesis revealed the role of TBC1D3 in insulin/IGF-1 signaling pathway. We document here that TBC1D3 intensifies insulin/IGF-1-induced signal transduction through intricate, yet elegant fine-tuning of signaling mechanisms. We show that TBC1D3 expression substantially delayed ubiquitination and degradation of insulin receptor substrate-1 (IRS-1). This effect is achieved through suppression of serine phosphorylation at S636/639, S307 and S312 of IRS-1, which are key phosphorylation sites required for IRS-1 degradation. Furthermore, we report that the effect of TBC1D3 on IRS-1:S636/639 phosphorylation is mediated through TBC1D3-induced activation of protein phosphatase 2A (PP2A), followed by suppression of T389 phosphorylation on p70 S6 kinase (S6K). TBC1D3 specifically interacts with PP2A regulatory subunit B56γ, indicating that TBC1D3 and PP2A B56γ operate jointly to promote S6K:T389 dephosphorylation. These findings suggest that TBC1D3 plays an unanticipated and potentially unique role in the fine-tuning of insulin/IGF-1 signaling, while providing novel insights into the regulation of tumorigenesis by a hominoid-specific protein.


Subject(s)
GTPase-Activating Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , GTPase-Activating Proteins/metabolism , Humans , Phosphorylation , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins/metabolism
6.
Sci Signal ; 2(89): pe59, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19797272

ABSTRACT

The identification and characterization of human-specific genes and the cellular processes that the encoded proteins control have the potential to help us understand at the molecular level what makes humans different from other species. The sequencing of the human genome and the genomes of closely related primates has revealed the presence of a small number of human- or human-lineage-specific genes that have no orthologs in lower species. Human-specific and human-lineage-specific genes are likely to function as regulators of cell signaling events, and by fine-tuning pathways, the encoded proteins may contribute to human-specific characteristics and behaviors. In addition, human-specific genes may represent biomarkers for examining human-specific characteristics of various diseases. Investigation of the gene encoding TBC1D3 is one example of a search that may lead to understanding the evolution and the function of human-specific genes, because it is absent in lower species and present in high copy number in the human genome.


Subject(s)
Signal Transduction/genetics , Animals , Biological Evolution , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/physiology , Hominidae/genetics , Hominidae/physiology , Humans , Models, Genetic , Oncogene Proteins/genetics , Oncogene Proteins/physiology , Proto-Oncogene Proteins , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Species Specificity
7.
Parasitol Res ; 105(1): 47-55, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19205738

ABSTRACT

Previously, we showed that oleic acid (OA) induces Trypanosoma cruzi metacyclogenesis through a signaling pathway involving de novo diacylglycerol biosynthesis and simultaneous protein kinase C (PKC) activation. Herein, we demonstrated that OA also triggers a transient Ca(2+) signal in epimastigotes, necessary for parasite differentiation, that could account for PKC activation. In addition, we found that this free fatty acid (FFA) directly stimulated in vitro the activity of T. cruzi PKC in a dose-response way. We determined the presence of classical and novel PKC isoenzymes that were differentially expressed in the infective amastigotes (alpha and delta) and tripomastigotes (alpha, beta, and gamma) and in the non-infective epimastigotes (alpha, beta, gamma, and delta). We also demonstrated that OA induced in epimastigotes the translocation of PKC alpha, beta, gamma, and delta to the membrane, indicating a selective effect of this FFA. To establish a correlation between T. cruzi metacyclogenesis induced by OA and the activation of a particular PKC isoenzyme, the specific PKC inhibitors Ro 32-0432 and Rottlerin (9-30 nM and 5-35 microM, respectively) were employed. These compounds, even at the lowest concentrations assayed, abrogated both epimastigote differentiation and membrane translocation of PKC beta, gamma, and delta. These findings strongly support a key role for classical and novel PKC isoenzymes in the signaling pathways involved in T. cruzi metacyclogenesis induced by OA.


Subject(s)
Morphogenesis/drug effects , Oleic Acid/metabolism , Protein Kinase C/metabolism , Signal Transduction , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/growth & development , Amino Acid Sequence , Animals , Calcium/metabolism , Isoenzymes/metabolism , Molecular Sequence Data , Sequence Alignment
8.
J Biol Chem ; 283(19): 13233-42, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18319245

ABSTRACT

Hominoid- and human-specific genes may have evolved to modulate signaling pathways of a higher order of complexity. TBC1D3 is a hominoid-specific oncogene encoded by a cluster of eight paralogs on chromosome 17. Initial work indicates that TBC1D3 is widely expressed in human tissues ( Hodzic, D., Kong, C., Wainszelbaum, M. J., Charron, A. J., Su, X., and Stahl, P. D. (2006) Genomics 88, 731-736 ). In this study, we show that TBC1D3 expression has a powerful effect on cell proliferation that is further enhanced by epidermal growth factor (EGF) in both human and mouse cell lines. EGF activation of the Erk and protein kinase B/Akt pathways is enhanced, both in amplitude and duration, by TBC1D3 expression, whereas RNA interference silencing of TBC1D3 suppresses the activation. Light microscopy and Western blot experiments demonstrate that increased signaling in response to EGF is coupled with a significant delay in EGF receptor (EGFR) trafficking and degradation, which significantly extends the life span of EGFR. Moreover, TBC1D3 suppresses polyubiquitination of the EGFR and the recruitment of c-Cbl. Using the Ras binding domain of Raf1 to monitor GTP-Ras we show that TBC1D3 expression enhances Ras activation in quiescent cells, which is further increased by EGF treatment. We speculate that TBC1D3 may alter Ras GTP loading. We conclude that the expression of TBC1D3 generates a delay in EGFR degradation, a decrease in ubiquitination, and a failure to recruit adapter proteins that ultimately dysregulate EGFR signal transduction and enhance cell proliferation. Altered growth factor receptor trafficking and GTP-Ras turnover may be sites where recently evolved genes such as TBC1D3 selectively modulate signaling in hominoids and humans.


Subject(s)
ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , MAP Kinase Signaling System , Oncogene Proteins/metabolism , ras Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Proliferation/drug effects , Enzyme Activation , Epidermal Growth Factor/pharmacology , GTPase-Activating Proteins/genetics , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Proteins/genetics , Protein Binding , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Substrate Specificity , Ubiquitination
9.
Genomics ; 88(6): 731-736, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16863688

ABSTRACT

TBC1D3 is a member of the TBC1 domain family of proteins that stimulates the intrinsic GTPase activity of RAB5A, an essential actor in early endosome trafficking. Oncogenic properties of TBC1D3 have been demonstrated previously both in vitro and in mouse models. Although the oncogenic mechanism of TBC1D3 has yet to be elucidated, the TBC1D3 locus (chromosome 17q12) is amplified in 15% of primary prostate tumors. Here, we describe eight highly related TBC1D3 paralogues located within that genomic region, potentially encoding six variant TBC1D3 proteins. We found that human tissues display specific transcription patterns of these paralogues. Furthermore, that pattern was altered in several primary prostate tumors in comparison to healthy prostate tissues. Potential TBC1D3 oncogenic mechanisms are discussed in light of these results.


Subject(s)
Chromosomes, Human, Pair 17/genetics , GTPase-Activating Proteins/genetics , Gene Dosage , Genetic Variation , Oncogene Proteins/genetics , Oncogenes/genetics , Chromosome Mapping , Female , GTPase-Activating Proteins/metabolism , Humans , Male , Oncogene Proteins/metabolism , Organ Specificity , Placenta/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Exp Cell Res ; 312(12): 2238-51, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16650848

ABSTRACT

The endosomal compartment and the plasma membrane form a complex partnership that controls signal transduction and trafficking of different molecules. The specificity and functionality of the early endocytic pathway are regulated by a growing number of Rab GTPases, particularly Rab5. In this study, we demonstrate that IL4 (a Th-2 cytokine) and prostaglandin E2 (PGE2) synergistically induce Rab5 and several Rab effector proteins, including Rin1 and EEA1, and promote the formation of an enlarged early endocytic (EEE) compartment. Endosome enlargement is linked to a substantial induction of the mannose receptor (MR), a well-characterized macrophage endocytic receptor. Both MR levels and MR-mediated endocytosis are enhanced approximately 7-fold. Fluid-phase endocytosis is also elevated in treated cells. Light microscopy and fractionation studies reveal that MR colocalizes predominantly with Rab5a and partially with Rab11, an endosomal recycling pathway marker. Using retroviral expression of Rab5a:S34N, a dominant negative mutant, and siRNA Rab5a silencing, we demonstrate that Rab5a is essential for the large endosome phenotype and for localization of MR in these structures. We speculate that the EEE is maintained by activated Rab5, and that the EEE phenotype is part of some macrophage developmental program such as cell fusion, a characteristic of IL4-stimulated cells.


Subject(s)
Dinoprostone/pharmacology , Endosomes/drug effects , Interleukin-4/pharmacology , Macrophages/drug effects , rab5 GTP-Binding Proteins/metabolism , Animals , Autoantigens/metabolism , Carrier Proteins/metabolism , Drug Synergism , Endocytosis/drug effects , Endosomes/physiology , Endosomes/ultrastructure , Intracellular Signaling Peptides and Proteins , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Macrophages/metabolism , Macrophages/physiology , Male , Mannans/metabolism , Mannose Receptor , Mannose-Binding Lectins/analysis , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Pinocytosis/drug effects , RNA, Small Interfering/genetics , Receptors, Cell Surface/analysis , Receptors, Cell Surface/metabolism , Vesicular Transport Proteins , rab GTP-Binding Proteins/analysis , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/analysis , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins , ras GTPase-Activating Proteins/metabolism
11.
Biochem J ; 375(Pt 3): 705-12, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12887332

ABSTRACT

Intestinal extracts of Triatoma infestans induce cell differentiation of Trypanosoma cruzi epimastigotes into the infective metacyclic form. Part of this effect can be explained by the presence of haemoglobin fragments, which stimulate trypanosomal adenylate cyclase. In this work we examined the metacyclogenic activity of lipids present in this intestinal extract. We found that lipid extracts of the intestinal extract have significant stimulatory effects that reside with the free-fatty-acid fraction, especially oleic acid. These compounds stimulate de novo diacylglycerol formation and protein kinase C activity in the parasite. Moreover, metacyclogenesis is stimulated by phorbol esters and cell-permeant diacylglycerol, while protein kinase C down-regulation or incubation with inhibitors of this kinase abrogates this effect. These results indicate that free fatty acids are a novel signal, inducing metacyclogenesis, acting through a pathway involving diacylglycerol biosynthesis and protein kinase C activation.


Subject(s)
Cell Differentiation/drug effects , Fatty Acids, Nonesterified/pharmacology , Sulfonamides , Trypanosoma cruzi/drug effects , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Bucladesine/pharmacology , Cell Division/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diglycerides/biosynthesis , Diglycerides/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Nonesterified/pharmacokinetics , Intestines/chemistry , Isoquinolines/pharmacology , Oleic Acid/pharmacokinetics , Oleic Acid/pharmacology , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Triatoma/chemistry , Trypanosoma cruzi/cytology , Trypanosoma cruzi/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...