Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Clin Cancer Res ; 29(6): 1086-1101, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36355054

ABSTRACT

PURPOSE: We evaluated the activity of AZD8205, a B7-H4-directed antibody-drug conjugate (ADC) bearing a novel topoisomerase I inhibitor (TOP1i) payload, alone and in combination with the PARP1-selective inhibitor AZD5305, in preclinical models. EXPERIMENTAL DESIGN: IHC and deep-learning-based image analysis algorithms were used to assess prevalence and intratumoral heterogeneity of B7-H4 expression in human tumors. Several TOP1i-ADCs, prepared with Val-Ala or Gly-Gly-Phe-Gly peptide linkers, with or without a PEG8 spacer, were compared in biophysical, in vivo efficacy, and rat toxicology studies. AZD8205 mechanism of action and efficacy studies were conducted in human cancer cell line and patient-derived xenograft (PDX) models. RESULTS: Evaluation of IHC-staining density on a per-cell basis revealed a range of heterogeneous B7-H4 expression across patient tumors. This informed selection of bystander-capable Val-Ala-PEG8-TOP1i payload AZ14170133 and development of AZD8205, which demonstrated improved stability, efficacy, and safety compared with other linker-payload ADCs. In a study of 26 PDX tumors, single administration of 3.5 mg/kg AZD8205 provided a 69% overall response rate, according to modified RECIST criteria, which correlated with homologous recombination repair (HRR) deficiency (HRD) and elevated levels of B7-H4 in HRR-proficient models. Addition of AZD5305 sensitized very low B7-H4-expressing tumors to AZD8205 treatment, independent of HRD status and in models representing clinically relevant mechanisms of PARPi resistance. CONCLUSIONS: These data provide evidence for the potential utility of AZD8205 for treatment of B7-H4-expressing tumors and support the rationale for an ongoing phase 1 clinical study (NCT05123482). See related commentary by Pommier and Thomas, p. 991.


Subject(s)
Immunoconjugates , Neoplasms , Rats , Humans , Animals , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Topoisomerase I Inhibitors , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly (ADP-Ribose) Polymerase-1/genetics
2.
Anal Chem ; 94(3): 1795-1803, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35005896

ABSTRACT

Gemcitabine (dFdC) is a common treatment for pancreatic cancer; however, it is thought that treatment may fail because tumor stroma prevents drug distribution to tumor cells. Gemcitabine is a pro-drug with active metabolites generated intracellularly; therefore, visualizing the distribution of parent drug as well as its metabolites is important. A multimodal imaging approach was developed using spatially coregistered mass spectrometry imaging (MSI), imaging mass cytometry (IMC), multiplex immunofluorescence microscopy (mIF), and hematoxylin and eosin (H&E) staining to assess the local distribution and metabolism of gemcitabine in tumors from a genetically engineered mouse model of pancreatic cancer (KPC) allowing for comparisons between effects in the tumor tissue and its microenvironment. Mass spectrometry imaging (MSI) enabled the visualization of the distribution of gemcitabine (100 mg/kg), its phosphorylated metabolites dFdCMP, dFdCDP and dFdCTP, and the inactive metabolite dFdU. Distribution was compared to small-molecule ATR inhibitor AZD6738 (25 mg/kg), which was codosed. Gemcitabine metabolites showed heterogeneous distribution within the tumor, which was different from the parent compound. The highest abundance of dFdCMP, dFdCDP, and dFdCTP correlated with distribution of endogenous AMP, ADP, and ATP in viable tumor cell regions, showing that gemcitabine active metabolites are reaching the tumor cell compartment, while AZD6738 was located to nonviable tumor regions. The method revealed that the generation of active, phosphorylated dFdC metabolites as well as treatment-induced DNA damage primarily correlated with sites of high proliferation in KPC PDAC tumor tissue, rather than sites of high parent drug abundance.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Mice , Multimodal Imaging , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Gemcitabine
3.
Cancer Res ; 82(6): 1140-1152, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35078817

ABSTRACT

AZD6738 (ceralasertib) is a potent and selective orally bioavailable inhibitor of ataxia telangiectasia and Rad3-related (ATR) kinase. ATR is activated in response to stalled DNA replication forks to promote G2-M cell-cycle checkpoints and fork restart. Here, we found AZD6738 modulated CHK1 phosphorylation and induced ATM-dependent signaling (pRAD50) and the DNA damage marker γH2AX. AZD6738 inhibited break-induced replication and homologous recombination repair. In vitro sensitivity to AZD6738 was elevated in, but not exclusive to, cells with defects in the ATM pathway or that harbor putative drivers of replication stress such as CCNE1 amplification. This translated to in vivo antitumor activity, with tumor control requiring continuous dosing and free plasma exposures, which correlated with induction of pCHK1, pRAD50, and γH2AX. AZD6738 showed combinatorial efficacy with agents associated with replication fork stalling and collapse such as carboplatin and irinotecan and the PARP inhibitor olaparib. These combinations required optimization of dose and schedules in vivo and showed superior antitumor activity at lower doses compared with that required for monotherapy. Tumor regressions required at least 2 days of daily dosing of AZD6738 concurrent with carboplatin, while twice daily dosing was required following irinotecan. In a BRCA2-mutant patient-derived triple-negative breast cancer (TNBC) xenograft model, complete tumor regression was achieved with 3 to5 days of daily AZD6738 per week concurrent with olaparib. Increasing olaparib dosage or AZD6738 dosing to twice daily allowed complete tumor regression even in a BRCA wild-type TNBC xenograft model. These preclinical data provide rationale for clinical evaluation of AZD6738 as a monotherapy or combinatorial agent. SIGNIFICANCE: This detailed preclinical investigation, including pharmacokinetics/pharmacodynamics and dose-schedule optimizations, of AZD6738/ceralasertib alone and in combination with chemotherapy or PARP inhibitors can inform ongoing clinical efforts to treat cancer with ATR inhibitors.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/metabolism , Carboplatin , Humans , Indoles , Irinotecan , Morpholines/pharmacology , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Sulfoxides/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
4.
EBioMedicine ; 68: 103396, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34049239

ABSTRACT

BACKGROUND: Chemotherapy and targeted agent anti-cancer efficacy is largely dependent on the proliferative state of tumours, as exemplified by agents that target DNA synthesis/replication or mitosis. As a result, cell cycle specificities of a number of cancer drugs are well known. However, they are yet to be described in a quantifiable manner. METHODS: A scalable cell synchronisation protocol used to screen a library of 235 anti-cancer compounds exposed over six hours in G1 or S/G2 accumulated AsPC-1 cells to generate a cell cycle specificity (CCS) score. FINDINGS: The synchronisation method was associated with reduced method-related cytotoxicity compared to nocodazole, delivering sufficient cell cycle purity and cell numbers to run high-throughput drug library screens. Compounds were identified with G1 and S/G2-associated specificities that, overall, functionally matched with a compound's target/mechanism of action. This annotation was used to describe a synergistic schedule using the CDK4/6 inhibitor, palbociclib, prior to gemcitabine/AZD6738 as well as describe the correlation between the CCS score and published synergistic/antagonistic drug schedules. INTERPRETATION: This is the first highly quantitative description of cell cycle-dependent drug sensitivities that utilised a tractable and tolerated method with potential uses outside the present study. Drug treatments such as those shown to be G1 or S/G2 associated may benefit from scheduling considerations such as after CDK4/6 inhibitors and being first in drug sequences respectively. FUNDING: Cancer Research UK (CRUK) Institute core grants C14303/A17197 and C9545/A29580. The Li Ka Shing Centre where this work was performed was generously funded by CK Hutchison Holdings Limited, the University of Cambridge, CRUK, The Atlantic Philanthropies and others.


Subject(s)
Deoxycytidine/analogs & derivatives , Neoplasms/metabolism , Nocodazole/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Tubulin Modulators/pharmacology , Cell Culture Techniques , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , HeLa Cells , High-Throughput Screening Assays , Humans , MCF-7 Cells , Neoplasms/drug therapy , Time Factors , Gemcitabine
5.
Cancers (Basel) ; 13(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540838

ABSTRACT

Kinase-targeted agents demonstrate antitumor activity in advanced metastatic clear cell renal cell carcinoma (ccRCC), which remains largely incurable. Integration of genomic approaches through small-molecules and genetically based high-throughput screening holds the promise of improved discovery of candidate targets for cancer therapy. The 786-O cell line represents a model for most ccRCC that have a loss of functional pVHL (von Hippel-Lindau). A multiplexed assay was used to study the cellular fitness of a panel of engineered ccRCC isogenic 786-O VHL- cell lines in response to a collection of targeted cancer therapeutics including kinase inhibitors, allowing the interrogation of over 2880 drug-gene pairs. Among diverse patterns of drug sensitivities, investigation of the mechanistic effect of one selected drug combination on tumor spheroids and ex vivo renal tumor slice cultures showed that VHL-defective ccRCC cells were more vulnerable to the combined inhibition of the CK2 and ATM kinases than wild-type VHL cells. Importantly, we found that HIF-2α acts as a key mediator that potentiates the response to combined CK2/ATM inhibition by triggering ROS-dependent apoptosis. Importantly, our findings reveal a selective killing of VHL-deficient renal carcinoma cells and provide a rationale for a mechanism-based use of combined CK2/ATM inhibitors for improved patient care in metastatic VHL-ccRCC.

6.
Br J Cancer ; 123(9): 1424-1436, 2020 10.
Article in English | MEDLINE | ID: mdl-32741974

ABSTRACT

BACKGROUND: Personalised medicine strategies may improve outcomes in pancreatic ductal adenocarcinoma (PDAC), but validation of predictive biomarkers is required. Having developed a clinical trial to assess the ATR inhibitor, AZD6738, in combination with gemcitabine (ATRi/gem), we investigated ATM loss as a predictive biomarker of response to ATRi/gem in PDAC. METHODS: Through kinase inhibition, siRNA depletion and CRISPR knockout of ATM, we assessed how ATM targeting affected the sensitivity of PDAC cells to ATRi/gem. Using flow cytometry, immunofluorescence and immunoblotting, we investigated how ATRi/gem synergise in ATM-proficient and ATM-deficient cells, before assessing the impact of ATM loss on ATRi/gem sensitivity in vivo. RESULTS: Complete loss of ATM function (through pharmacological inhibition or CRISPR knockout), but not siRNA depletion, sensitised to ATRi/gem. In ATM-deficient cells, ATRi/gem-induced replication catastrophe was augmented, while phospho-Chk2-T68 and phospho-KAP1-S824 persisted via DNA-PK activity. ATRi/gem caused growth delay in ATM-WT xenografts in NSG mice and induced regression in ATM-KO xenografts. CONCLUSIONS: ATM loss augments replication catastrophe-mediated cell death induced by ATRi/gem and may predict clinical responsiveness to this combination. ATM status should be carefully assessed in tumours from patients with PDAC, since distinction between ATM-low and ATM-null could be critical in maximising the success of clinical trials using ATM expression as a predictive biomarker.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Sulfoxides/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/physiology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Synergism , Female , Gene Knockout Techniques , Humans , Indoles , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Morpholines , Pancreatic Neoplasms/pathology , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Quinolines/administration & dosage , RNA, Small Interfering/pharmacology , Sulfonamides , Sulfoxides/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
7.
Mol Cancer Ther ; 17(8): 1670-1682, 2018 08.
Article in English | MEDLINE | ID: mdl-29891488

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers, and overall survival rates have barely improved over the past five decades. The antimetabolite gemcitabine remains part of the standard of care but shows very limited antitumor efficacy. Ataxia telangiectasia and Rad3-related protein (ATR), the apical kinase of the intra-S-phase DNA damage response, plays a central role in safeguarding cells from replication stress and can therefore limit the efficacy of antimetabolite drug therapies. We investigated the ability of the ATR inhibitor, AZD6738, to prevent the gemcitabine-induced intra-S-phase checkpoint activation and evaluated the antitumor potential of this combination in vitro and in vivo In PDAC cell lines, AZD6738 inhibited gemcitabine-induced Chk1 activation, prevented cell-cycle arrest, and restrained RRM2 accumulation, leading to the strong induction of replication stress markers only with the combination. Moreover, synergistic growth inhibition was identified in a panel of 5 mouse and 7 human PDAC cell lines using both Bliss Independence and Loewe models. In clonogenic assays, the combination abrogated survival at concentrations for which single agents had minor effects. In vivo, AZD6738 in combination with gemcitabine was well tolerated and induced tumor regression in a subcutaneous allograft model of a KrasG12D; Trp53R172H; Pdx-Cre (KPC) mouse cancer cell line, significantly extending survival. Remarkably, the combination also induced regression of a subgroup of KPC autochthonous tumors, which generally do not respond well to conventional chemotherapy. Altogether, our data suggest that AZD6738 in combination with gemcitabine merits evaluation in a clinical trial in patients with PDAC. Mol Cancer Ther; 17(8); 1670-82. ©2018 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Pyrimidines/therapeutic use , Sulfoxides/therapeutic use , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Humans , Indoles , Mice , Morpholines , Pyrimidines/pharmacology , Sulfonamides , Sulfoxides/pharmacology , Gemcitabine
8.
Cancer Res ; 78(11): 3054-3066, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29735549

ABSTRACT

Combination of cytotoxic therapy with emerging DNA damage response inhibitors (DDRi) has been limited by tolerability issues. However, the goal of most combination trials has been to administer DDRi with standard-of-care doses of chemotherapy. We hypothesized that mechanism-guided treatment scheduling could reduce the incidence of dose-limiting toxicities and enable tolerable multitherapeutic regimens. Integrative analyses of mathematical modeling and single-cell assays distinguished the synergy kinetics of WEE1 inhibitor (WEE1i) from CHEK1 inhibitor (CHK1i) by potency, spatiotemporal perturbation, and mitotic effects when combined with gemcitabine. These divergent properties collectively supported a triple-agent strategy, whereby a pulse of gemcitabine and CHK1i followed by WEE1i durably suppressed tumor cell growth. In xenografts, CHK1i exaggerated replication stress without mitotic CDK hyperactivation, enriching a geminin-positive subpopulation and intratumoral gemcitabine metabolite. Without overt toxicity, addition of WEE1i to low-dose gemcitabine and CHK1i was most effective in tumor control compared with single and double agents. Overall, our work provides quantitative insights into the mechanisms of DDRi chemosensitization, leading to the rational development of a tolerable multitherapeutic regimen.Significance: Multiple lines of mechanistic insight regarding DNA damage response inhibitors rationally guide the preclinical development of a tolerable multitherapeutic regimen.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/11/3054/F1.large.jpg Cancer Res; 78(11); 3054-66. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Replication/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays/methods , Gemcitabine
9.
Sci Rep ; 8(1): 5, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311668

ABSTRACT

Pathogenic bacteria induce eukaryotic cell damage which range from discrete modifications of signalling pathways, to morphological alterations and even to cell death. Accurate quantitative detection of these events is necessary for studying host-pathogen interactions and for developing strategies to protect host organisms from bacterial infections. Investigation of morphological changes is cumbersome and not adapted to high-throughput and kinetics measurements. Here, we describe a simple and cost-effective method based on automated analysis of live cells with stained nuclei, which allows real-time quantification of bacteria-induced eukaryotic cell damage at single-cell resolution. We demonstrate that this automated high-throughput microscopy approach permits screening of libraries composed of interference-RNA, bacterial strains, antibodies and chemical compounds in ex vivo infection settings. The use of fluorescently-labelled bacteria enables the concomitant detection of changes in bacterial growth. Using this method named CLIQ-BID (Cell Live Imaging Quantification of Bacteria Induced Damage), we were able to distinguish the virulence profiles of different pathogenic bacterial species and clinical strains.


Subject(s)
Bacterial Physiological Phenomena , Eukaryotic Cells/microbiology , Eukaryotic Cells/pathology , Molecular Imaging/methods , Animals , Endothelial Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , NIH 3T3 Cells
10.
ACS Med Chem Lett ; 8(7): 726-731, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28740606

ABSTRACT

ERK is the effector kinase of the RAS-RAF-MEK-ERK signaling cascade, which promotes cell transformation and malignancy in many cancers and is thus a major drug target in oncology. Kinase inhibitors targeting RAF or MEK are already used for the treatment of certain cancers, such as melanoma. Although the initial response to these drugs can be dramatic, development of drug resistance is a major challenge, even with combination therapies targeting both RAF and MEK. Importantly, most resistance mechanisms still rely on activation of the downstream effector kinase ERK, making it a promising target for drug development efforts. Here, we report the design and structural/functional characterization of a set of bivalent ERK inhibitors that combine a small molecule inhibitor that binds to the ATP-binding pocket with a peptide that selectively binds to an ERK protein interaction surface, the D-site recruitment site (DRS). Our studies show that the lead bivalent inhibitor, SBP3, has markedly improved potency compared to the small molecule inhibitor alone. Unexpectedly, we found that SBP3 also binds to several ERK-related kinases that contain a DRS, highlighting the importance of experimentally verifying the predicted specificity of bivalent inhibitors. However, SBP3 does not target any other kinases belonging to the same CMGC branch of the kinome. Additionally, our modular click chemistry inhibitor design facilitates the generation of different combinations of small molecule inhibitors with ERK-targeting peptides.

11.
Cell Mol Life Sci ; 72(17): 3305-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25990538

ABSTRACT

Structurally, protein kinase CK2 consists of two catalytic subunits (α and α') and two regulatory subunits (ß), which play a critical role in targeting specific CK2 substrates. Compelling evidence shows the complexity of the CK2 cellular signaling network and supports the view that this enzyme is a key component of regulatory protein kinase networks that are involved in several aspects of cancer. CK2 both activates and suppresses the expression of a number of essential oncogenes and tumor suppressors, and its expression and activity are upregulated in blood tumors and virtually all solid tumors. The prognostic significance of CK2α expression in association with various clinicopathological parameters highlighted this kinase as an adverse prognostic marker in breast cancer. In addition, several recent studies reported its implication in the regulation of the epithelial-to-mesenchymal transition (EMT), an early step in cancer invasion and metastasis. In this review, we briefly overview the contribution of CK2 to several aspects of cancer and discuss how in mammary epithelial cells, the expression of its CK2ß regulatory subunit plays a critical role in maintaining an epithelial phenotype through CK2-mediated control of key EMT-related transcription factors. Importantly, decreased CK2ß expression in breast tumors is correlated with inefficient phosphorylation and nuclear translocation of Snail1 and Foxc2, ultimately leading to EMT induction. This review highlights the pivotal role played by CK2ß in the mammary epithelial phenotype and discusses how a modest alteration in its expression may be sufficient to induce dramatic effects facilitating the early steps in tumor cell dissemination through the coordinated regulation of two key transcription factors.


Subject(s)
Breast Neoplasms/enzymology , Casein Kinase II/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/genetics , Signal Transduction/physiology , Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Forkhead Transcription Factors/metabolism , Humans , Snail Family Transcription Factors
12.
J Biol Chem ; 289(15): 10431-10444, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24584939

ABSTRACT

Most breast cancers are estrogen receptor-positive and treated with antiestrogens, but aberrant signaling networks can induce drug resistance. One of these networks involves the scaffolding protein BCAR1/p130CAS, which regulates cell growth and migration/invasion. A less investigated scaffolding protein that also confers antiestrogen resistance is the SH2 domain-containing protein BCAR3. BCAR1 and BCAR3 bind tightly to each other through their C-terminal domains, thus potentially connecting their associated signaling networks. However, recent studies using BCAR1 and BCAR3 interaction mutants concluded that association between the two proteins is not critical for many of their interrelated activities regulating breast cancer malignancy. We report that these previously used BCAR mutations fail to cause adequate loss-of-function of the complex. By using structure-based BCAR1 and BCAR3 mutants that lack the ability to interact, we show that BCAR3-induced antiestrogen resistance in MCF7 breast cancer cells critically depends on its ability to bind BCAR1. Interaction with BCAR3 increases the levels of phosphorylated BCAR1, ultimately potentiating BCAR1-dependent antiestrogen resistance. Furthermore, antiestrogen resistance in cells overexpressing BCAR1/BCAR3 correlates with increased ERK1/2 activity. Inhibiting ERK1/2 through overexpression of the regulatory protein PEA15 negates the resistance, revealing a key role for ERK1/2 in BCAR1/BCAR3-induced antiestrogen resistance. Reverse-phase protein array data show that PEA15 levels in invasive breast cancers correlate with patient survival, suggesting that PEA15 can override ERK1/2 activation by BCAR1/BCAR3 and other upstream regulators. We further uncovered that the BCAR3-related NSP3 can also promote antiestrogen resistance. Thus, strategies to disrupt BCAR1-BCAR3/NSP3 complexes and associated signaling networks could ultimately lead to new breast cancer therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Crk-Associated Substrate Protein/metabolism , Estrogen Receptor Modulators/pharmacology , Estrogens/pharmacology , Signal Transduction , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors , HEK293 Cells , Humans , Lentivirus/genetics , MCF-7 Cells , Microscopy, Fluorescence , Mutation , Phenotype , Phosphorylation , Plasmids/metabolism , Protein Conformation
13.
Nat Commun ; 4: 1681, 2013.
Article in English | MEDLINE | ID: mdl-23575685

ABSTRACT

ERK1/2 kinases are the principal effectors of a central signalling cascade that converts extracellular stimuli into cell proliferation and migration responses and, when deregulated, can promote cell oncogenic transformation. The scaffolding protein PEA-15 is a death effector domain protein that directly interacts with ERK1/2 and affects ERK1/2 subcellular localization and phosphorylation. Here, to understand this ERK1/2 signalling complex, we have solved the crystal structures of PEA-15 bound to three different ERK2 phospho-conformers. The structures reveal that PEA-15 uses a bipartite binding mode, occupying two key docking sites of ERK2. Remarkably, PEA-15 can efficiently bind the ERK2 activation loop in the critical Thr-X-Tyr region in different phosphorylation states. PEA-15 binding triggers an extended allosteric conduit in dually phosphorylated ERK2, disrupting key features of active ERK2. At the same time PEA-15 binding protects ERK2 from dephosphorylation, thus setting the stage for immediate ERK activity upon its release from the PEA-15 inhibitory complex.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid
14.
Genes Cancer ; 3(5-6): 382-93, 2012 May.
Article in English | MEDLINE | ID: mdl-23226576

ABSTRACT

The CAS (CRK-associated substrate) family of adaptor proteins comprises 4 members, which share a conserved modular domain structure that enables multiple protein-protein interactions, leading to the assembly of intracellular signaling platforms. Besides their physiological role in signal transduction downstream of a variety of cell surface receptors, CAS proteins are also critical for oncogenic transformation and cancer cell malignancy through associations with a variety of regulatory proteins and downstream effectors. Among the regulatory partners, the 3 recently identified adaptor proteins constituting the NSP (novel SH2-containing protein) family avidly bind to the conserved carboxy-terminal focal adhesion-targeting (FAT) domain of CAS proteins. NSP proteins use an anomalous nucleotide exchange factor domain that lacks catalytic activity to form NSP-CAS signaling modules. Additionally, the NSP SH2 domain can link NSP-CAS signaling assemblies to tyrosine-phosphorylated cell surface receptors. NSP proteins can potentiate CAS function by affecting key CAS attributes such as expression levels, phosphorylation state, and subcellular localization, leading to effects on cell adhesion, migration, and invasion as well as cell growth. The consequences of these activities are well exemplified by the role that members of both families play in promoting breast cancer cell invasiveness and resistance to antiestrogens. In this review, we discuss the intriguing interplay between the NSP and CAS families, with a particular focus on cancer signaling networks.

15.
Nat Struct Mol Biol ; 18(12): 1381-7, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081014

ABSTRACT

Members of the novel SH2-containing protein (NSP) and Crk-associated substrate (Cas) protein families form multidomain signaling platforms that mediate cell migration and invasion through a collection of distinct signaling motifs. Members of each family interact via their respective C-terminal domains, but the mechanism of this association has remained enigmatic. Here we present the crystal structures of the C-terminal domain from the NSP protein BCAR3 and the complex of NSP3 with p130Cas. BCAR3 adopts the Cdc25-homology fold of Ras GTPase exchange factors, but it has a 'closed' conformation incapable of enzymatic activity. The structure of the NSP3-p130Cas complex reveals that this closed conformation is instrumental for interaction of NSP proteins with a focal adhesion-targeting domain present in Cas proteins. This enzyme-to-adaptor conversion enables high-affinity, yet promiscuous, interactions between NSP and Cas proteins and represents an unprecedented mechanistic paradigm linking cellular signaling networks.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Crk-Associated Substrate Protein/chemistry , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Conserved Sequence , Crk-Associated Substrate Protein/metabolism , Guanine Nucleotide Exchange Factors , Humans , Models, Molecular , Protein Structure, Tertiary
16.
J Neurosci ; 30(39): 13201-10, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20881139

ABSTRACT

Shep1 is a multidomain signaling protein that forms a complex with Cas, a key scaffolding component of integrin signaling pathways, to promote the migration of non-neuronal cells. However, the physiological function of Shep1 in the nervous system remains unknown. Interestingly, we found that Shep1 and Cas are both concentrated in the axons of developing olfactory sensory neurons (OSNs). These neurons extend their axons from the olfactory epithelium to the olfactory bulb located at the anterior tip of the forebrain. However, in developing Shep1 knock-out mice, we did not detect penetration of OSN axons across the pial basement membrane surrounding the olfactory bulb, suggesting that Shep1 function is important for the establishment of OSN connections with the olfactory bulb. Interestingly, we observed reduced levels of Cas tyrosine phosphorylation in OSN axons of Shep1 knock-out mice, suggesting compromised Cas signaling function. Indeed, when embedded in a three-dimensional gel of basement membrane proteins, explants from Shep1 knock-out olfactory epithelium extend neuronal processes less efficiently than explants from control epithelium. Furthermore, ectopic expression of Shep1 in non-neuronal cells promotes cell migration through a collagen gel. Later in development, loss of Shep1 function also causes a marked reduction in olfactory bulb size and disruption of bulb lamination, which may be primarily attributable to the defective innervation. The greatly reduced OSN connections and hypoplasia of the olfactory bulb, likely resulting in anosmia, are reminiscent of the symptoms of Kallmann syndrome, a human developmental disease that can be caused by mutations in a growing number of genes.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Movement/physiology , Growth Cones/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Olfactory Receptor Neurons/physiology , Prosencephalon/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Movement/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism
17.
Proc Natl Acad Sci U S A ; 107(44): 18944-9, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956287

ABSTRACT

The marginal zone is a cellular niche bordering the marginal sinus of the spleen that contains specialized B-cell and macrophage subsets poised to capture bloodborne antigens. Marginal zone B cells are retained in this niche by integrin-mediated signaling induced by G protein-coupled receptors (GPCRs) and, likely, the B-cell receptor (BCR). Sphingosine-1-phosphate (S1P) signaling via the S1P family of GPCRs is known to be essential for B-cell localization in the marginal zone, but little is known about the downstream signaling events involved. Here, we demonstrate that the adaptor protein SHEP1 is required for marginal zone B-cell maturation. SHEP1 functions in concert with the scaffolding protein CasL, because we show that SHEP1 and CasL are constitutively associated in B cells. SHEP1 association is required for the BCR or S1P receptor(s) to induce the conversion of CasL into its serine/threonine hyperphosphorylated form, which is important for lymphocyte adhesion and motility. Thus, SHEP1 orchestrates marginal zone B-cell movement and retention as a key downstream effector of the BCR and S1P receptors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/metabolism , Cell Movement/physiology , Signal Transduction/physiology , Spleen/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/cytology , Lysophospholipids/genetics , Lysophospholipids/metabolism , Mice , Mice, Knockout , Phosphorylation/physiology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/genetics , Sphingosine/metabolism , Spleen/cytology
18.
Cell Signal ; 22(11): 1745-52, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20603213

ABSTRACT

The members of the p130Cas (Cas) family are important scaffolding proteins that orchestrate cell adhesion, migration and invasiveness downstream of integrin adhesion receptors and receptor tyrosine kinases by recruiting enzymes and structural molecules. Shep1, BCAR3/AND-34 and NSP1 define a recently identified family of SH2 domain-containing proteins that constitutively bind Cas proteins through a Cdc25-type nucleotide exchange factor-like domain. To gain insight into the functional interplay between Shep1 and Cas in vivo, we have inactivated the Shep1 gene in the mouse through Cre-mediated deletion of the exon encoding the SH2 domain. Analysis of Cas tyrosine phosphorylation in the brains of newborn mice, where Shep1 is highly expressed, revealed a strong decrease in Cas substrate domain phosphorylation in knockout compared to wild-type brains. Src family kinases bind to Cas via their SH3 and SH2 domains, which contributes to their activation, and phosphorylate multiple tyrosines in the Cas substrate domain. These tyrosine-phosphorylated motifs represent docking sites for the Crk adaptor, linking Cas to the downstream Rac1 and Rap1 GTPases to regulate cell adhesion and actin cytoskeleton organization. Accordingly, we detected lower Cas-Crk association and lower phosphorylation of the Src activation loop in Shep1 knockout brains compared to wild-type. Conversely, Shep1 transfection in COS cells increases Cas tyrosine phosphorylation. The SH2 domain is likely critical for the effects of Shep1 on Cas and Src signaling because the knockout mice express Shep1 fragments that lack the amino-terminal region including the SH2 domain, presumably due to aberrant translation from internal ATG codons. These fragments retain the ability to increase Cas levels in transfected cells, similar to full-length Shep1. However, they do not affect Cas phosphorylation on their own or in the presence of co-transfected full-length Shep1. They also do not show dominant negative effects on the activity of full-length Shep1 in vivo because the heterozygous mice, which express the fragments, have a normal life span. This is in contrast to the homozygous knockout mice, most of which die soon after birth. These data demonstrate that Shep1 plays a critical role in the in vivo regulation of Src activity and Cas downstream signaling through Crk, and suggest that the SH2 domain of Shep1 is critical for these effects.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Crk-Associated Substrate Protein/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-crk/metabolism , rac1 GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/metabolism , src Homology Domains , src-Family Kinases/metabolism
19.
Biochim Biophys Acta ; 1778(3): 794-809, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17961505

ABSTRACT

Endothelial cells lining the vessel wall are connected by adherens, tight and gap junctions. These junctional complexes are related to those found at epithelial junctions but with notable changes in terms of specific molecules and organization. Endothelial junctional proteins play important roles in tissue integrity but also in vascular permeability, leukocyte extravasation and angiogenesis. In this review, we will focus on specific mechanisms of endothelial tight and adherens junctions.


Subject(s)
Adherens Junctions/physiology , Adherens Junctions/ultrastructure , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Tight Junctions/physiology , Tight Junctions/ultrastructure , Animals , Antigens, CD/chemistry , Antigens, CD/physiology , Cadherins/chemistry , Cadherins/physiology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/physiology , Cell Proliferation , Contact Inhibition , Homeostasis , Humans , Inflammation/pathology , Inflammation/physiopathology , Membrane Proteins/chemistry , Membrane Proteins/physiology , Models, Biological , Models, Molecular , Multiprotein Complexes , Neovascularization, Physiologic , Signal Transduction , beta Catenin/chemistry , beta Catenin/physiology
20.
Trends Cardiovasc Med ; 16(2): 55-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16473763

ABSTRACT

Because angiogenesis is a key step in a number of pathologic processes, including tumor growth and atherosclerosis, many research studies have investigated the regulatory signals active at various stages of vascular invasion. The differential activities of the endothelial junction protein vascular endothelial (VE)-cadherin reflect the versatile behavior of endothelial cells between vascular quiescence and angiogenesis. VE-cadherin function and signaling are deeply modified in proliferating cells, and this conversion is accompanied by phosphorylation of the protein on tyrosine residues and enhanced transcription of its gene. Recent advances in the complex interplay between protein tyrosine kinases and phosphatases regulating VE-cadherin phosphorylation and function are discussed in this review.


Subject(s)
Antigens, CD/physiology , Atherosclerosis/pathology , Cadherins/physiology , Endothelium, Vascular/physiology , Neovascularization, Pathologic/etiology , Antigens, CD/genetics , Antigens, CD/metabolism , Atherosclerosis/etiology , Cadherins/genetics , Cadherins/metabolism , Capillary Permeability , Cell Cycle/physiology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Gene Expression Regulation/physiology , Humans , Neoplasms/pathology , Phosphorylation , Tyrosine/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...