94 results on '"MacBeath G"'
Search Results
2. Heteromeric tandem IgG4/IgG1 Fc
- Author
-
Casaletto, J.B., primary, Geddie, M.L., additional, Abu-Yousif, A.O., additional, Masson, K., additional, Fulgham, A., additional, Boudot, A., additional, Maiwald, T., additional, Kearns, J.D., additional, Kohli, N., additional, Su, S., additional, Razlog, M., additional, Raue, A., additional, Kalra, A., additional, Hakansson, M., additional, Logan, D.T., additional, Welin, M., additional, Chattopadhyay, S., additional, Harms, B.D., additional, Nielsen, U.B., additional, Schoeberl, B., additional, Lugovskoy, A.A., additional, and MacBeath, G., additional
- Published
- 2019
- Full Text
- View/download PDF
3. Crystal structure of EpCAM in complex with scFv
- Author
-
Casaletto, J.B., primary, Geddie, M.L., additional, Abu-Yousif, A.O., additional, Masson, K., additional, Fulgham, A., additional, Boudot, A., additional, Maiwald, T., additional, Kearns, J.D., additional, Kohli, N., additional, Su, S., additional, Razlog, M., additional, Raue, A., additional, Kalra, A., additional, Hakansson, M., additional, Logan, D.T., additional, Welin, M., additional, Chattopadhyay, S., additional, Harms, B.D., additional, Nielsen, U.B., additional, Schoeberl, B., additional, Lugovskoy, A.A., additional, and MacBeath, G., additional
- Published
- 2019
- Full Text
- View/download PDF
4. Crystal structure of Sema domain of the Met receptor in complex with FAB
- Author
-
Casaletto, J.B., primary, Geddie, M.L., additional, Abu-Yousif, A.O., additional, Masson, K., additional, Fulgham, A., additional, Boudot, A., additional, Maiwald, T., additional, Kearns, J.D., additional, Kohli, N., additional, Su, S., additional, Razlog, M., additional, Raue, A., additional, Kalra, A., additional, Hakansson, M., additional, Logan, D.T., additional, Welin, M., additional, Chattopadhyay, S., additional, Harms, B.D., additional, Nielsen, U.B., additional, Schoeberl, B., additional, Lugovskoy, A.A., additional, and MacBeath, G., additional
- Published
- 2019
- Full Text
- View/download PDF
5. ABP-100: A tetravalent bispecific T-cell engaging antibody for HER2+ solid tumors
- Author
-
Boudot, A., primary, Huang, X., additional, Murphy, S., additional, Appleman, V., additional, Xu, H., additional, Cheung, N.-K., additional, and Macbeath, G., additional
- Published
- 2018
- Full Text
- View/download PDF
6. The oligoclonal anti-EGFR antibody MM-151 synergizes with trametinib in KRAS wildtype and mutant colorectal cancer models resistant to conventional monoclonal anti-EGFR antibodies
- Author
-
Lahdenranta, J., primary, Gerami-Moayed, N., additional, Burenkova, O., additional, Masson, K., additional, Raue, A., additional, Wainszelbaum, M., additional, and MacBeath, G., additional
- Published
- 2016
- Full Text
- View/download PDF
7. 1170P - ABP-100: A tetravalent bispecific T-cell engaging antibody for HER2+ solid tumors
- Author
-
Boudot, A., Huang, X., Murphy, S., Appleman, V., Xu, H., Cheung, N.-K., and Macbeath, G.
- Published
- 2018
- Full Text
- View/download PDF
8. 438 MM-131: A bispecific antibody that inhibits c-Met signaling through avid binding to the EpCAM tumor antigen
- Author
-
Harms, B.D., primary, Lugovskoy, A., additional, Abu-Yousif, A., additional, Fulgham, A., additional, Geddie, M., additional, Su, S.V., additional, Kohli, N., additional, Johnson, B., additional, Masson, K., additional, Nielsen, U.B., additional, Schoeberl, B., additional, and MacBeath, G., additional
- Published
- 2014
- Full Text
- View/download PDF
9. A Meta-Analysis of Biomarkers in Three Randomized, Phase 2 Studies of Mm-121, a Ligand-Blocking Anti-Erbb3 Antibody, in Patients with Ovarian, Lung, and Breast Cancers
- Author
-
Macbeath, G., primary, Adiwijaya, B., additional, Liu, J., additional, Sequist, L.V., additional, Pujade-Lauraine, E., additional, Higgins, M., additional, Tabah-Fisch, I., additional, Pearlberg, J., additional, Moyo, V., additional, Kubasek, W., additional, Nering, R., additional, and Czibere, A., additional
- Published
- 2014
- Full Text
- View/download PDF
10. 398 - The oligoclonal anti-EGFR antibody MM-151 synergizes with trametinib in KRAS wildtype and mutant colorectal cancer models resistant to conventional monoclonal anti-EGFR antibodies
- Author
-
Lahdenranta, J., Gerami-Moayed, N., Burenkova, O., Masson, K., Raue, A., Wainszelbaum, M., and MacBeath, G.
- Published
- 2016
- Full Text
- View/download PDF
11. Abstract P1-07-03: Quantification of HER2 expression at the single cell level and HER2 intratumoral heterogeneity of breast cancer tissue samples using automated image analysis
- Author
-
Geretti, E, primary, Paragas, V, additional, Onsum, M, additional, Kudla, A, additional, Moulis, S, additional, Luus, L, additional, Wickham, T, additional, McDonagh, C, additional, MacBeath, G, additional, and Hendriks, B, additional
- Published
- 2012
- Full Text
- View/download PDF
12. 141 MM-121, an Anti-ErbB3 Antibody, Inhibits PI3K/AKT Signaling and Viability in Platinum-resistant Ovarian Cells and in Primary Ascites Derived From Chemo-resistant Ovarian Cancer Patients
- Author
-
Curley, M., primary, Kalra, A., additional, Fulgham, A., additional, Xiao, D., additional, Allen, J., additional, Wainszelbaum, M., additional, Garcia, G., additional, Kubasek, W., additional, and MacBeath, G., additional
- Published
- 2012
- Full Text
- View/download PDF
13. Profiling phospho-signaling networks in breast cancer using reverse-phase protein arrays
- Author
-
Gujral, T S, primary, Karp, R L, additional, Finski, A, additional, Chan, M, additional, Schwartz, P E, additional, MacBeath, G, additional, and Sorger, P, additional
- Published
- 2012
- Full Text
- View/download PDF
14. C60-1, a PDZ domain designed using statistical coupling analysis
- Author
-
Larson, C., primary, Stiffler, M., additional, Li, P., additional, Rosen, M., additional, MacBeath, G., additional, and Ranganathan, R., additional
- Published
- 2008
- Full Text
- View/download PDF
15. 246P - A Meta-Analysis of Biomarkers in Three Randomized, Phase 2 Studies of Mm-121, a Ligand-Blocking Anti-Erbb3 Antibody, in Patients with Ovarian, Lung, and Breast Cancers
- Author
-
Macbeath, G., Adiwijaya, B., Liu, J., Sequist, L.V., Pujade-Lauraine, E., Higgins, M., Tabah-Fisch, I., Pearlberg, J., Moyo, V., Kubasek, W., Nering, R., and Czibere, A.
- Published
- 2014
- Full Text
- View/download PDF
16. Cloning and analysis of a constitutive heat shock (cognate) protein 70 gene inducible by L-glutamine.
- Author
-
LéJohn, H.B., primary, Cameron, L.E., additional, Yang, B., additional, MacBeath, G., additional, Barker, D.S., additional, and Williams, S.A., additional
- Published
- 1994
- Full Text
- View/download PDF
17. Hydrolytic antibodies: variations on a theme
- Author
-
MacBeath, G
- Published
- 1996
- Full Text
- View/download PDF
18. A call to adapt the regulation of HLA testing for T cell receptor-based therapeutics.
- Author
-
Meyer M, Mahr A, Brewer J, Daniel V, Dell'Aringa J, Goldstone T, Hersey S, Johnston I, Larson P, Loveridge M, MacBeath G, Moyer M, Nagorsen D, Papa S, Peiser L, Ranade K, Rizzi R, Roers A, Schendel D, Sivakumar P, Tran E, Türeci Ö, Weigand L, Wennborg A, Williams D, Yee C, and Britten CM
- Subjects
- Humans, Receptors, Antigen, T-Cell, Immunotherapy standards, Histocompatibility Testing
- Published
- 2024
- Full Text
- View/download PDF
19. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy.
- Author
-
Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, Tadros J, Ferretti AP, Liao S, Gurer C, Chen YH, Criscitiello S, Ricker CA, Dionne D, Rozenblatt-Rosen O, Uppaluri R, Haddad RI, Ashenberg O, Regev A, Van Allen EM, MacBeath G, Schoenfeld JD, and Wucherpfennig KW
- Subjects
- CD8-Positive T-Lymphocytes, Humans, Immunotherapy, Lymphocytes, Tumor-Infiltrating, Neoadjuvant Therapy, Tumor Microenvironment, Neoplasms therapy, Programmed Cell Death 1 Receptor
- Abstract
Neoadjuvant immune checkpoint blockade has shown promising clinical activity. Here, we characterized early kinetics in tumor-infiltrating and circulating immune cells in oral cancer patients treated with neoadjuvant anti-PD-1 or anti-PD-1/CTLA-4 in a clinical trial (NCT02919683). Tumor-infiltrating CD8 T cells that clonally expanded during immunotherapy expressed elevated tissue-resident memory and cytotoxicity programs, which were already active prior to therapy, supporting the capacity for rapid response. Systematic target discovery revealed that treatment-expanded tumor T cell clones in responding patients recognized several self-antigens, including the cancer-specific antigen MAGEA1. Treatment also induced a systemic immune response characterized by expansion of activated T cells enriched for tumor-infiltrating T cell clonotypes, including both pre-existing and emergent clonotypes undetectable prior to therapy. The frequency of activated blood CD8 T cells, notably pre-treatment PD-1-positive KLRG1-negative T cells, was strongly associated with intra-tumoral pathological response. These results demonstrate how neoadjuvant checkpoint blockade induces local and systemic tumor immunity., Competing Interests: Declaration of interests K.W.W. serves on the SAB of SQZ Biotech, Nextechinvest, Bisou Bioscience Company, and T-Scan Therapeutics and receives sponsored research funding from Novartis. He is a scientific co-founder of Immunitas Therapeutics. J.D.S. reports research support paid to the institution from Merck, BMS, Regeneron, Debiopharm; Consulting/Scientific Advisory Board/Travel fees: Genentech, Immunitas, Debiopharm, BMS, Nanobiotix, Tilos, Castle Biosciences, Astra Zeneca, LEK, Catenion, ACI Clinical, Astellas, Stimit, and Merck KGA; Expert witness fees. Stock options: Immunitas; Equity: Doximity. E.M.V.A. reports Advisory/Consulting: Tango Therapeutics, Genome Medical, Invitae, Enara Bio, Janssen, Manifold Bio, and Monte Rosa; Research support: Novartis, BMS; Equity: Tango Therapeutics, Genome Medical, Syapse, Enara Bio, Manifold Bio, Microsoft, and Monte Rosa; Travel reimbursement: Roche/Genentech; Patents: Institutional patents filed on chromatin mutations and immunotherapy response, and methods for clinical interpretation; intermittent legal consulting on patents for Foaley & Hoag. A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics, and until August 31, 2020, was an SAB member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov, and Thermo Fisher Scientific. From August 1, 2020, A.R. is an employee of Genentech and has equity in Roche., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Phase 1 dose escalation study of seribantumab (MM-121), an anti-HER3 monoclonal antibody, in patients with advanced solid tumors.
- Author
-
Denlinger CS, Keedy VL, Moyo V, MacBeath G, and Shapiro GI
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Dose-Response Relationship, Drug, Female, Half-Life, Humans, Male, Maximum Tolerated Dose, Middle Aged, Receptor, ErbB-3 antagonists & inhibitors, Antibodies, Monoclonal, Humanized pharmacokinetics, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Neoplasms drug therapy
- Abstract
Background Overactivation of human epidermal growth factor receptor 3 (HER3) triggers multiple intracellular pathways resulting in tumor cell survival. This Phase 1 study assessed the safety, efficacy, and pharmacokinetics (PK) of seribantumab, a fully human anti-HER3 monoclonal antibody. Methods Adult patients with advanced or refractory solid tumors were treated in six dose cohorts of seribantumab: 3.2, 6, 10, 15, or 20 mg/kg weekly, or 40 mg/kg loading dose followed by 20 mg/kg weekly maintenance dose (40/20 mg/kg) using a modified 3 + 3 dose escalation strategy with cohort expansion. Primary objectives were identification of a recommended Phase 2 dose (RP2D) and determination of objective response rate. Secondary objectives were assessment of safety, dose-limiting toxicities, and PK. Results Forty-four patients (26 dose escalation; 18 dose expansion) were enrolled. Seribantumab monotherapy was well tolerated with most adverse events being transient and mild to moderate (grade 1 or 2) in severity; maximum tolerated dose was not reached. The highest dose, 40/20 mg/kg, was identified as RP2D. Best response was stable disease, reported in 24% and 39% of patients during the dose escalation and expansion portions of the study, respectively. Seribantumab terminal half-life was ≈100 h; steady state concentrations were reached after 3-4 weekly doses. Conclusions Seribantumab monotherapy was well tolerated across all dose levels. Safety and PK data from this study support further seribantumab investigations in genomically defined populations.Clinical trial registration NCT00734305. August 12, 2008., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Phenotype, specificity and avidity of antitumour CD8 + T cells in melanoma.
- Author
-
Oliveira G, Stromhaug K, Klaeger S, Kula T, Frederick DT, Le PM, Forman J, Huang T, Li S, Zhang W, Xu Q, Cieri N, Clauser KR, Shukla SA, Neuberg D, Justesen S, MacBeath G, Carr SA, Fritsch EF, Hacohen N, Sade-Feldman M, Livak KJ, Boland GM, Ott PA, Keskin DB, and Wu CJ
- Subjects
- CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Datasets as Topic, Gene Expression Regulation, Humans, Lymphocytes, Tumor-Infiltrating immunology, Melanoma blood, Phenotype, Receptors, Antigen, T-Cell immunology, Single-Cell Analysis, Transcriptome genetics, Tumor Microenvironment, CD8-Positive T-Lymphocytes immunology, Melanoma immunology, Substrate Specificity immunology
- Abstract
Interactions between T cell receptors (TCRs) and their cognate tumour antigens are central to antitumour immune responses
1-3 ; however, the relationship between phenotypic characteristics and TCR properties is not well elucidated. Here we show, by linking the antigenic specificity of TCRs and the cellular phenotype of melanoma-infiltrating lymphocytes at single-cell resolution, that tumour specificity shapes the expression state of intratumoural CD8+ T cells. Non-tumour-reactive T cells were enriched for viral specificities and exhibited a non-exhausted memory phenotype, whereas melanoma-reactive lymphocytes predominantly displayed an exhausted state that encompassed diverse levels of differentiation but rarely acquired memory properties. These exhausted phenotypes were observed both among clonotypes specific for public overexpressed melanoma antigens (shared across different tumours) or personal neoantigens (specific for each tumour). The recognition of such tumour antigens was provided by TCRs with avidities inversely related to the abundance of cognate targets in melanoma cells and proportional to the binding affinity of peptide-human leukocyte antigen (HLA) complexes. The persistence of TCR clonotypes in peripheral blood was negatively affected by the level of intratumoural exhaustion, and increased in patients with a poor response to immune checkpoint blockade, consistent with chronic stimulation mediated by residual tumour antigens. By revealing how the quality and quantity of tumour antigens drive the features of T cell responses within the tumour microenvironment, we gain insights into the properties of the anti-melanoma TCR repertoire., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
22. Unbiased Screens Show CD8 + T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein.
- Author
-
Ferretti AP, Kula T, Wang Y, Nguyen DMV, Weinheimer A, Dunlap GS, Xu Q, Nabilsi N, Perullo CR, Cristofaro AW, Whitton HJ, Virbasius A, Olivier KJ Jr, Buckner LR, Alistar AT, Whitman ED, Bertino SA, Chattopadhyay S, and MacBeath G
- Subjects
- Adult, Aged, Betacoronavirus isolation & purification, COVID-19, Convalescence, Coronavirus immunology, Coronavirus Infections diagnosis, Coronavirus Nucleocapsid Proteins, Epitope Mapping, Epitopes, T-Lymphocyte, Female, Humans, Immunodominant Epitopes, Immunologic Memory, Male, Middle Aged, Nucleocapsid Proteins immunology, Pandemics, Phosphoproteins, Pneumonia, Viral diagnosis, Polyproteins, SARS-CoV-2, Viral Proteins immunology, Young Adult, Betacoronavirus immunology, CD8-Positive T-Lymphocytes immunology, Coronavirus Infections immunology, Pneumonia, Viral immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
Developing effective strategies to prevent or treat coronavirus disease 2019 (COVID-19) requires understanding the natural immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We used an unbiased, genome-wide screening technology to determine the precise peptide sequences in SARS-CoV-2 that are recognized by the memory CD8
+ T cells of COVID-19 patients. In total, we identified 3-8 epitopes for each of the 6 most prevalent human leukocyte antigen (HLA) types. These epitopes were broadly shared across patients and located in regions of the virus that are not subject to mutational variation. Notably, only 3 of the 29 shared epitopes were located in the spike protein, whereas most epitopes were located in ORF1ab or the nucleocapsid protein. We also found that CD8+ T cells generally do not cross-react with epitopes in the four seasonal coronaviruses that cause the common cold. Overall, these findings can inform development of next-generation vaccines that better recapitulate natural CD8+ T cell immunity to SARS-CoV-2., Competing Interests: Declaration of Interests A.P.F., Y.W., D.M.V.N., A.W., G.S.D., Q.X., N.N., C.R.P., A.W.C., H.J.W., A.V., K.J.O., S.A.B., and G.M. are employees of TScan Therapeutics. T.K. is a founder of TScan Therapeutics, and T.K. and G.M. hold equity in TScan Therapeutics. S.C. is a consultant for TScan Therapeutics. A patent application relating to the T-Scan technology used in the current manuscript was filed previously by The Brigham and Women’s Hospital, Inc. Provisional patent applications covering SARS-CoV-2 epitope sequences and TCR sequences described in the current manuscript as well as related uses, diagnostics, therapeutics, and vaccines for COVID-19 were filed by TScan Therapeutics, Inc., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
23. MM-131, a bispecific anti-Met/EpCAM mAb, inhibits HGF-dependent and HGF-independent Met signaling through concurrent binding to EpCAM.
- Author
-
Casaletto JB, Geddie ML, Abu-Yousif AO, Masson K, Fulgham A, Boudot A, Maiwald T, Kearns JD, Kohli N, Su S, Razlog M, Raue A, Kalra A, Håkansson M, Logan DT, Welin M, Chattopadhyay S, Harms BD, Nielsen UB, Schoeberl B, Lugovskoy AA, and MacBeath G
- Subjects
- Animals, Cell Line, Tumor, Epithelial Cell Adhesion Molecule metabolism, Humans, Mice, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Proto-Oncogene Proteins c-met metabolism, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacology, Antineoplastic Agents, Immunological pharmacology, Epithelial Cell Adhesion Molecule antagonists & inhibitors, Hepatocyte Growth Factor metabolism, Neoplasms, Experimental drug therapy, Proto-Oncogene Proteins c-met antagonists & inhibitors, Signal Transduction drug effects
- Abstract
Activation of the Met receptor tyrosine kinase, either by its ligand, hepatocyte growth factor (HGF), or via ligand-independent mechanisms, such as MET amplification or receptor overexpression, has been implicated in driving tumor proliferation, metastasis, and resistance to therapy. Clinical development of Met-targeted antibodies has been challenging, however, as bivalent antibodies exhibit agonistic properties, whereas monovalent antibodies lack potency and the capacity to down-regulate Met. Through computational modeling, we found that the potency of a monovalent antibody targeting Met could be dramatically improved by introducing a second binding site that recognizes an unrelated, highly expressed antigen on the tumor cell surface. Guided by this prediction, we engineered MM-131, a bispecific antibody that is monovalent for both Met and epithelial cell adhesion molecule (EpCAM). MM-131 is a purely antagonistic antibody that blocks ligand-dependent and ligand-independent Met signaling by inhibiting HGF binding to Met and inducing receptor down-regulation. Together, these mechanisms lead to inhibition of proliferation in Met-driven cancer cells, inhibition of HGF-mediated cancer cell migration, and inhibition of tumor growth in HGF-dependent and -independent mouse xenograft models. Consistent with its design, MM-131 is more potent in EpCAM-high cells than in EpCAM-low cells, and its potency decreases when EpCAM levels are reduced by RNAi. Evaluation of Met, EpCAM, and HGF levels in human tumor samples reveals that EpCAM is expressed at high levels in a wide range of Met-positive tumor types, suggesting a broad opportunity for clinical development of MM-131., Competing Interests: Conflict of interest statement: Several authors are or were employees of Merrimack Pharmaceuticals, Inc. at the time of contributing to the manuscript.
- Published
- 2019
- Full Text
- View/download PDF
24. Estimation of immune cell content in tumour tissue using single-cell RNA-seq data.
- Author
-
Schelker M, Feau S, Du J, Ranu N, Klipp E, MacBeath G, Schoeberl B, and Raue A
- Subjects
- Algorithms, Cells, Cultured, Humans, Immune System immunology, Immune System pathology, Neoplasms immunology, Neoplasms pathology, Stromal Cells metabolism, Tumor Microenvironment genetics, Gene Expression Profiling methods, Immune System metabolism, Neoplasms genetics, Single-Cell Analysis methods
- Abstract
As interactions between the immune system and tumour cells are governed by a complex network of cell-cell interactions, knowing the specific immune cell composition of a solid tumour may be essential to predict a patient's response to immunotherapy. Here, we analyse in depth how to derive the cellular composition of a solid tumour from bulk gene expression data by mathematical deconvolution, using indication-specific and cell type-specific reference gene expression profiles (RGEPs) from tumour-derived single-cell RNA sequencing data. We demonstrate that tumour-derived RGEPs are essential for the successful deconvolution and that RGEPs from peripheral blood are insufficient. We distinguish nine major cell types, as well as three T cell subtypes. Using the tumour-derived RGEPs, we can estimate the content of many tumours associated immune and stromal cell types, their therapeutically relevant ratios, as well as an improved gene expression profile of the malignant cells.
- Published
- 2017
- Full Text
- View/download PDF
25. Predicting ligand-dependent tumors from multi-dimensional signaling features.
- Author
-
Hass H, Masson K, Wohlgemuth S, Paragas V, Allen JE, Sevecka M, Pace E, Timmer J, Stelling J, MacBeath G, Schoeberl B, and Raue A
- Abstract
Targeted therapies have shown significant patient benefit in about 5-10% of solid tumors that are addicted to a single oncogene. Here, we explore the idea of ligand addiction as a driver of tumor growth. High ligand levels in tumors have been shown to be associated with impaired patient survival, but targeted therapies have not yet shown great benefit in unselected patient populations. Using an approach of applying Bagged Decision Trees (BDT) to high-dimensional signaling features derived from a computational model, we can predict ligand dependent proliferation across a set of 58 cell lines. This mechanistic, multi-pathway model that features receptor heterodimerization, was trained on seven cancer cell lines and can predict signaling across two independent cell lines by adjusting only the receptor expression levels for each cell line. Interestingly, for patient samples the predicted tumor growth response correlates with high growth factor expression in the tumor microenvironment, which argues for a co-evolution of both factors in vivo.
- Published
- 2017
- Full Text
- View/download PDF
26. A phase 1 study combining the HER3 antibody seribantumab (MM-121) and cetuximab with and without irinotecan.
- Author
-
Cleary JM, McRee AJ, Shapiro GI, Tolaney SM, O'Neil BH, Kearns JD, Mathews S, Nering R, MacBeath G, Czibere A, Sharma S, and Korn WM
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols pharmacology, Camptothecin adverse effects, Camptothecin pharmacology, Camptothecin therapeutic use, ErbB Receptors genetics, Female, Humans, Irinotecan, Male, Maximum Tolerated Dose, Middle Aged, Treatment Outcome, Young Adult, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Camptothecin analogs & derivatives, Cetuximab adverse effects, Cetuximab pharmacology, Cetuximab therapeutic use, Receptor, ErbB-3 immunology
- Abstract
Background HER3/EGFR heterodimers have been implicated as a mode of resistance to EGFR-directed therapies. Methods This Phase 1 trial assessed the tolerability, maximum tolerated dose (MTD) and pharmacokinetic (PK) properties of the HER-3 antibody seribantumab in combination with cetuximab (Part I) or cetuximab and irinotecan (Part II) in patients with EGFR-dependent cancers. In Part I, escalating doses of seribantumab and cetuximab were administered. In Part II of the trial, escalating doses of seribantumab/cetuximab were combined with irinotecan 180 mg/m2 administered every two weeks. Results 34 patients were enrolled in Part I (seribantumab/cetuximab) and 14 patients were enrolled in Part II (seribantumab/cetuximab/irinotecan). Common toxicities of seribantumab/cetuximab included acneiform rash, diarrhea, stomatitis, and paronychia. The MTD of Part I was seribantumab 40 mg/kg bolus, then 20 mg/kg weekly combined with cetuximab 400 mg/m2 bolus, then 250 mg/m2 IV weekly. Common toxicities reported in the seribantumab/cetuximab/irinotecan combination were similar to the Part I portion. However, toxicities were more frequent and severe with the triplet combination. There was one treatment-related death in Part II secondary to Grade 4 neutropenia and grade 3 diarrhea. Other dose-limiting toxicities in Part II were Grade 3 mucositis and Grade 3 diarrhea. A cholangiocarcinoma patient, previously untreated with EGFR-directed therapy, had a confirmed partial response (PR). One colorectal cancer patient, previously treated with EGFR-directed therapy, had an unconfirmed PR. Conclusions Seribantumab/cetuximab was well tolerated and patients experienced toxicities typical to EGFR inhibition. Unlike the seribantumab/cetuximab doublet, seribantumab/cetuximab/irinotecan was difficult to tolerate in this heavily pretreated population. There was limited efficacy of the combination therapy.
- Published
- 2017
- Full Text
- View/download PDF
27. Systems biology driving drug development: from design to the clinical testing of the anti-ErbB3 antibody seribantumab (MM-121).
- Author
-
Schoeberl B, Kudla A, Masson K, Kalra A, Curley M, Finn G, Pace E, Harms B, Kim J, Kearns J, Fulgham A, Burenkova O, Grantcharova V, Yarar D, Paragas V, Fitzgerald J, Wainszelbaum M, West K, Mathews S, Nering R, Adiwijaya B, Garcia G, Kubasek B, Moyo V, Czibere A, Nielsen UB, and MacBeath G
- 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., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
28. Randomized Phase II Trial of Seribantumab in Combination With Paclitaxel in Patients With Advanced Platinum-Resistant or -Refractory Ovarian Cancer.
- Author
-
Liu JF, Ray-Coquard I, Selle F, Poveda AM, Cibula D, Hirte H, Hilpert F, Raspagliesi F, Gladieff L, Harter P, Siena S, Del Campo JM, Tabah-Fisch I, Pearlberg J, Moyo V, Riahi K, Nering R, Kubasek W, Adiwijaya B, Czibere A, Naumann RW, Coleman RL, Vergote I, MacBeath G, and Pujade-Lauraine E
- Subjects
- Aged, Antibodies, Monoclonal, Humanized, Disease-Free Survival, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Humans, Kaplan-Meier Estimate, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Staging, Ovarian Neoplasms mortality, Ovarian Neoplasms pathology, Prognosis, Proportional Hazards Models, Prospective Studies, Risk Assessment, Survival Analysis, Treatment Outcome, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Biomarkers, Tumor analysis, Ovarian Neoplasms drug therapy, Paclitaxel administration & dosage
- Abstract
Purpose Seribantumab is a fully human immunoglobulin G2 monoclonal antibody that binds to human epidermal growth factor receptor (HER) 3 (ErbB3), blocking heregulin (HRG) -mediated ErbB3 signaling and inducing ErbB3 receptor downregulation. This open-label randomized phase II study evaluated progression-free survival (PFS) with seribantumab in combination with once-per-week paclitaxel compared with paclitaxel alone in patients with platinum-resistant or -refractory ovarian cancer. A key secondary objective was to determine if any of five prespecified biomarkers predicted benefit from seribantumab. Patients and Methods Patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal cancer were randomly assigned at a ratio of two to one to receive seribantumab plus paclitaxel or paclitaxel alone. Patients underwent pretreatment core needle biopsy; archival tumor samples were also obtained to support biomarker analyses. Results A total of 223 patients were randomly assigned (seribantumab plus paclitaxel, n = 140; paclitaxel alone, n = 83). Median PFS in the unselected intent-to-treat population was 3.75 months with seribantumab plus paclitaxel compared with 3.68 months with paclitaxel alone (hazard ratio [HR], 1.027; 95% CI, 0.741 to 1.425; P = .864). Among patients whose tumors had detectable HRG mRNA and low HER2 (n = 57 [38%] of 151 with available biomarker data), increased treatment benefit was observed in those receiving seribantumab plus paclitaxel compared with paclitaxel alone (PFS HR, 0.37; 95% CI, 0.18 to 0.76; P = .007). The HR in patients not meeting these criteria was 1.80 (95% CI, 1.08 to 2.98; P = .023). Conclusion The addition of seribantumab to paclitaxel did not result in improved PFS in unselected patients. Exploratory analyses suggest that detectable HRG and low HER2, biomarkers that link directly to the mechanism of action of seribantumab, identified patients who might benefit from this combination. Future clinical trials are needed to validate this finding and should preselect for HRG expression and focus on cancers with low HER2 levels.
- Published
- 2016
- Full Text
- View/download PDF
29. Seribantumab, an Anti-ERBB3 Antibody, Delays the Onset of Resistance and Restores Sensitivity to Letrozole in an Estrogen Receptor-Positive Breast Cancer Model.
- Author
-
Curley MD, Sabnis GJ, Wille L, Adiwijaya BS, Garcia G, Moyo V, Kazi AA, Brodie A, and MacBeath G
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Humanized, Antineoplastic Agents pharmacology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Female, Humans, Immunoblotting, Letrozole, Mice, Inbred BALB C, Mice, Nude, Neuregulin-1 pharmacology, Ovariectomy, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation drug effects, Receptor, ErbB-3 immunology, Receptor, ErbB-3 metabolism, Receptors, Estrogen metabolism, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism, Antibodies, Monoclonal pharmacology, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Nitriles pharmacology, Receptor, ErbB-3 antagonists & inhibitors, Triazoles pharmacology, Xenograft Model Antitumor Assays
- Abstract
Heregulin-driven ERBB3 signaling has been implicated as a mechanism of resistance to cytotoxic and antiendocrine therapies in preclinical breast cancer models. In this study, we evaluated the effects of seribantumab (MM-121), a heregulin-blocking anti-ERBB3 monoclonal antibody, alone and in combination with the aromatase inhibitor letrozole, on cell signaling and tumor growth in a preclinical model of postmenopausal estrogen receptor-positive (ER(+)) breast cancer. In vitro, heregulin treatment induced estrogen receptor phosphorylation in MCF-7Ca cells, and long-term letrozole-treated (LTLT-Ca) cells had increased expression and activation levels of EGFR, HER2, and ERBB3. Treatment with seribantumab, but not letrozole, inhibited basal and heregulin-mediated ERBB receptor phosphorylation and downstream effector activation in letrozole-sensitive (MCF-7Ca) and -refractory (LTLT-Ca) cells. Notably, in MCF-7Ca-derived xenograft tumors, cotreatment with seribantumab and letrozole had increased antitumor activity compared with letrozole alone, which was accompanied by downregulated PI3K/MTOR signaling both prior to and after the development of resistance to letrozole. Moreover, the addition of an MTOR inhibitor to this treatment regimen did not improve antitumor activity and was not well tolerated. Our results demonstrate that heregulin-driven ERBB3 signaling mediates resistance to letrozole in a preclinical model of ER(+) breast cancer, suggesting that heregulin-expressing ER(+) breast cancer patients may benefit from the addition of seribantumab to antiendocrine therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
30. Heregulin and HER3 are prognostic biomarkers in oropharyngeal squamous cell carcinoma.
- Author
-
Qian G, Jiang N, Wang D, Newman S, Kim S, Chen Z, Garcia G, MacBeath G, Shin DM, Khuri FR, Chen ZG, and Saba NF
- Subjects
- Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Cyclin-Dependent Kinase Inhibitor p16, ErbB Receptors metabolism, Genetic Markers genetics, Head and Neck Neoplasms genetics, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Humans, Neoplasm Proteins genetics, Prognosis, Receptor, ErbB-2 metabolism, Receptor, ErbB-3 genetics, Retrospective Studies, Squamous Cell Carcinoma of Head and Neck, Survival Analysis, Carcinoma, Squamous Cell mortality, Head and Neck Neoplasms mortality, Neuregulin-1 genetics, Receptor, ErbB-3 metabolism
- Abstract
Background: Although heregulin and human epidermal growth factor receptor 3 (HER3) are frequently expressed at high levels in patients with head and neck cancer, their prognostic value remains unclear. The authors explored the prognostic significance of heregulin/HER3 expression in patients with oropharyngeal squamous cell carcinoma (OPSCC), taking into account other HER family members as well as p16 status., Methods: Ninety-six primary tumor specimens from patients with OPSCC were retrospectively collected and analyzed for heregulin messenger RNA (mRNA) using in situ hybridization and for HER3, epidermal growth factor receptor, and human epidermal growth factor receptor 2 (HER2) using quantitative immunohistochemistry. Heregulin and HER3 mRNA levels were also examined among different tumor types using The Cancer Genome Atlas database., Results: High heregulin mRNA (> the median) correlated significantly with poor overall survival (OS) (hazard ratio [HR], 8.48; 95% confidence interval [95% CI], 2.17-33.17 [P =.002]) but not disease-free survival (HR, 1.52; 95% CI, 0.64-3.65 [P =.341]) in patients with OPSCC. Heregulin mRNA correlated negatively with OS in both patients with p16-positive (P =.049) and p16-negative (P =.091) OPSCC on univariate analysis. High HER3 (> the median) also correlated with poor OS (HR, 4.68; 95% CI, 1.47-14.90 [P =.009]) on multivariate analysis. Epidermal growth factor receptor levels independently correlated with disease-free survival (P =.025) and inversely correlated with p16 status (P =.012). In addition, The Cancer Genome Atlas data demonstrated that head and neck squamous cell carcinoma exhibits higher heregulin expression compared with other solid tumor types examined., Conclusions: High heregulin mRNA and high HER3 protein levels were found to independently correlate with poor OS in patients with OPSCC. These data support targeting HER3 in patients with heregulin-high OPSCC and warrant further clinical investigation., (© 2015 American Cancer Society.)
- Published
- 2015
- Full Text
- View/download PDF
31. Heregulin-ErbB3-Driven Tumor Growth Persists in PI3 Kinase Mutant Cancer Cells.
- Author
-
Yarar D, Lahdenranta J, Kubasek W, Nielsen UB, and MacBeath G
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Cell Line, Tumor, Disease Models, Animal, Female, Gene Expression, Humans, Neoplasms pathology, Phosphatidylinositol 3-Kinases metabolism, Receptor, ErbB-3 antagonists & inhibitors, Receptor, ErbB-3 genetics, Signal Transduction, Xenograft Model Antitumor Assays, Mutation, Neoplasms genetics, Neoplasms metabolism, Neuregulin-1 metabolism, Phosphatidylinositol 3-Kinases genetics, Receptor, ErbB-3 metabolism
- Abstract
PI3K is frequently mutated in cancer and plays an important role in cell growth and survival. Heregulin (HRG)-mediated autocrine or paracrine signaling through the receptor tyrosine kinase ErbB3 potently activates the PI3K/AKT pathway and has been shown to mediate resistance to a wide variety of anticancer agents. Although PI3K functions downstream of HRG-ErbB3, it is unknown whether activating mutations in PI3K render HRG ineffective. If so, patients with PI3K mutations would not be expected to benefit from ErbB3-directed therapies. Here, we find that a subset of cell lines harboring activating PI3K mutations can be further growth-stimulated by HRG, and this effect is blocked by incubation with seribantumab (MM-121), a monoclonal anti-ErbB3 antibody. Although expression of mutant PI3K in wild-type PI3K cells frequently results in loss of HRG-stimulated growth, some cell lines continue to respond to HRG. In cell lines where HRG-stimulated growth is lost, this loss is invariably accompanied by a reduction in ErbB3 levels, a corresponding increase in basal phosphorylation levels of FOXO-family transcription factors, and a reduction in HRG-induced downstream signaling. Importantly, HRG-stimulated growth is partially rescued by re-expressing ErbB3. This response is blocked by seribantumab, indicating that ErbB3 levels rather than downstream signaling proteins limit HRG-stimulated growth in PI3K mutant cells. Overall, these results suggest that activating mutations in PI3K do not preclude potential benefit from ErbB3-directed therapy, but that it may be important to measure ErbB3 levels in patients with PI3K mutant cancers to determine if they would benefit., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
32. A multiscale statistical mechanical framework integrates biophysical and genomic data to assemble cancer networks.
- Author
-
AlQuraishi M, Koytiger G, Jenney A, MacBeath G, and Sorger PK
- Subjects
- Algorithms, Area Under Curve, False Positive Reactions, Genetic Variation, Genome, Human, Genomics, HEK293 Cells, Humans, Models, Statistical, Mutagenesis, Site-Directed, Mutation, ROC Curve, Receptor, IGF Type 1 genetics, src Homology Domains, Models, Genetic, Neoplasms genetics
- Abstract
Functional interpretation of genomic variation is critical to understanding human disease, but it remains difficult to predict the effects of specific mutations on protein interaction networks and the phenotypes they regulate. We describe an analytical framework based on multiscale statistical mechanics that integrates genomic and biophysical data to model the human SH2-phosphoprotein network in normal and cancer cells. We apply our approach to data in The Cancer Genome Atlas (TCGA) and test model predictions experimentally. We find that mutations mapping to phosphoproteins often create new interactions but that mutations altering SH2 domains result almost exclusively in loss of interactions. Some of these mutations eliminate all interactions, but many cause more selective loss, thereby rewiring specific edges in highly connected subnetworks. Moreover, idiosyncratic mutations appear to be as functionally consequential as recurrent mutations. By synthesizing genomic, structural and biochemical data, our framework represents a new approach to the interpretation of genetic variation.
- Published
- 2014
- Full Text
- View/download PDF
33. A noncanonical Frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis.
- Author
-
Gujral TS, Chan M, Peshkin L, Sorger PK, Kirschner MW, and MacBeath G
- Subjects
- Animals, Cell Line, Tumor, Heterografts, Humans, Mice, Nude, Neoplasm Metastasis pathology, Neoplasm Transplantation, STAT3 Transcription Factor metabolism, Wnt Proteins metabolism, Cell Movement, Epithelial-Mesenchymal Transition, Frizzled Receptors metabolism, Wnt Signaling Pathway
- Abstract
Wnt signaling plays a critical role in embryonic development, and genetic aberrations in this network have been broadly implicated in colorectal cancer. We find that the Wnt receptor Frizzled2 (Fzd2) and its ligands Wnt5a/b are elevated in metastatic liver, lung, colon, and breast cancer cell lines and in high-grade tumors and that their expression correlates with markers of epithelial-mesenchymal transition (EMT). Pharmacologic and genetic perturbations reveal that Fzd2 drives EMT and cell migration through a previously unrecognized, noncanonical pathway that includes Fyn and Stat3. A gene signature regulated by this pathway predicts metastasis and overall survival in patients. We have developed an antibody to Fzd2 that reduces cell migration and invasion and inhibits tumor growth and metastasis in xenografts. We propose that targeting this pathway could provide benefit for patients with tumors expressing high levels of Fzd2 and Wnt5a/b.
- Published
- 2014
- Full Text
- View/download PDF
34. Systematic analysis of bacterial effector-postsynaptic density 95/disc large/zonula occludens-1 (PDZ) domain interactions demonstrates Shigella OspE protein promotes protein kinase C activation via PDLIM proteins.
- Author
-
Yi CR, Allen JE, Russo B, Lee SY, Heindl JE, Baxt LA, Herrera BB, Kahoud E, MacBeath G, and Goldberg MB
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Bacterial Proteins chemistry, Conserved Sequence, Focal Adhesions metabolism, HEK293 Cells, HeLa Cells, Humans, Intracellular Space microbiology, Molecular Sequence Data, Mutant Proteins metabolism, Peptides chemistry, Peptides metabolism, Protein Array Analysis, Protein Binding, Saccharomyces cerevisiae metabolism, Shigella, Signal Transduction, Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing metabolism, Bacterial Proteins metabolism, Cytoskeletal Proteins chemistry, Cytoskeletal Proteins metabolism, LIM Domain Proteins chemistry, LIM Domain Proteins metabolism, Protein Interaction Domains and Motifs, Protein Kinase C metabolism
- Abstract
Diseases caused by many Gram-negative bacterial pathogens depend on the activities of bacterial effector proteins that are delivered into eukaryotic cells via specialized secretion systems. Effector protein function largely depends on specific subcellular targeting and specific interactions with cellular ligands. PDZ domains are common domains that serve to provide specificity in protein-protein interactions in eukaryotic systems. We show that putative PDZ-binding motifs are significantly enriched among effector proteins delivered into mammalian cells by certain bacterial pathogens. We use PDZ domain microarrays to identify candidate interaction partners of the Shigella flexneri effector proteins OspE1 and OspE2, which contain putative PDZ-binding motifs. We demonstrate in vitro and in cells that OspE proteins interact with PDLIM7, a member of the PDLIM family of proteins, which contain a PDZ domain and one or more LIM domains, protein interaction domains that participate in a wide variety of functions, including activation of isoforms of protein kinase C (PKC). We demonstrate that activation of PKC during S. flexneri infection is attenuated in the absence of PDLIM7 or OspE proteins and that the OspE PDZ-binding motif is required for wild-type levels of PKC activation. These results are consistent with a model in which binding of OspE to PDLIM7 during infection regulates the activity of PKC isoforms that bind to the PDLIM7 LIM domain., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
35. Combination of anti-HER3 antibody MM-121/SAR256212 and cetuximab inhibits tumor growth in preclinical models of head and neck squamous cell carcinoma.
- Author
-
Jiang N, Wang D, Hu Z, Shin HJ, Qian G, Rahman MA, Zhang H, Amin AR, Nannapaneni S, Wang X, Chen Z, Garcia G, MacBeath G, Shin DM, Khuri FR, Ma J, Chen ZG, and Saba NF
- Subjects
- Animals, Antibodies, Monoclonal immunology, Apoptosis drug effects, Apoptosis immunology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell immunology, Cell Growth Processes drug effects, Cell Growth Processes immunology, Cell Line, Tumor, Cetuximab, Combined Modality Therapy, Disease Models, Animal, Female, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms immunology, Humans, Immunohistochemistry, Mice, Mice, Nude, Random Allocation, Signal Transduction, Squamous Cell Carcinoma of Head and Neck, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Agents pharmacology, Carcinoma, Squamous Cell therapy, Head and Neck Neoplasms therapy, Receptor, ErbB-3 antagonists & inhibitors, Receptor, ErbB-3 immunology
- Abstract
The EGFR monoclonal antibody cetuximab is the only approved targeted agent for treating head and neck squamous cell carcinoma (HNSCC). Yet resistance to cetuximab has hindered its activity in this disease. Intrinsic or compensatory HER3 signaling may contribute to cetuximab resistance. To investigate the therapeutic benefit of combining MM-121/SAR256212, an anti-HER3 monoclonal antibody, with cetuximab in HNSCC, we initially screened 12 HNSCC cell lines for total and phosphorylated levels of the four HER receptors. We also investigated the combination of MM-121 with cetuximab in preclinical models of HNSCC. Our results revealed that HER3 is widely expressed and activated in HNSCC cell lines. MM-121 strongly inhibited phosphorylation of HER3 and AKT. When combined with cetuximab, MM-121 exerted a more potent antitumor activity through simultaneously inhibiting the activation of HER3 and EGFR and consequently the downstream PI3K/AKT and ERK pathways in vitro. Both high and low doses of MM-121 in combination with cetuximab significantly suppressed tumor growth in xenograft models and inhibited activations of HER3, EGFR, AKT, and ERK in vivo. Our work is the first report on this new combination in HNSCC and supports the concept that HER3 inhibition may play an important role in future therapy of HNSCC. Our results open the door for further mechanistic studies to better understand the role of HER3 in resistance to EGFR inhibitors in HNSCC., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
36. Single-cell quantitative HER2 measurement identifies heterogeneity and distinct subgroups within traditionally defined HER2-positive patients.
- Author
-
Onsum MD, Geretti E, Paragas V, Kudla AJ, Moulis SP, Luus L, Wickham TJ, McDonagh CF, MacBeath G, and Hendriks BS
- Subjects
- Animals, Breast Neoplasms classification, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Cluster Analysis, Female, Fluorescent Antibody Technique, Humans, Mice, Mice, Nude, Neoplasms pathology, Reference Standards, Reproducibility of Results, Stomach Neoplasms classification, Stomach Neoplasms metabolism, Stomach Neoplasms pathology, Tissue Array Analysis, Urinary Bladder Neoplasms classification, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Genetic Heterogeneity, Neoplasms classification, Neoplasms metabolism, Receptor, ErbB-2 metabolism, Single-Cell Analysis methods
- Abstract
Human epidermal growth factor receptor 2 (HER2) is an important biomarker for breast and gastric cancer prognosis and patient treatment decisions. HER2 positivity, as defined by IHC or fluorescent in situ hybridization testing, remains an imprecise predictor of patient response to HER2-targeted therapies. Challenges to correct HER2 assessment and patient stratification include intratumoral heterogeneity, lack of quantitative and/or objective assays, and differences between measuring HER2 amplification at the protein versus gene level. We developed a novel immunofluorescence method for quantitation of HER2 protein expression at the single-cell level on FFPE patient samples. Our assay uses automated image analysis to identify and classify tumor versus non-tumor cells, as well as quantitate the HER2 staining for each tumor cell. The HER2 staining level is converted to HER2 protein expression using a standard cell pellet array stained in parallel with the tissue sample. This approach allows assessment of HER2 expression and heterogeneity within a tissue section at the single-cell level. By using this assay, we identified distinct subgroups of HER2 heterogeneity within traditional definitions of HER2 positivity in both breast and gastric cancers. Quantitative assessment of intratumoral HER2 heterogeneity may offer an opportunity to improve the identification of patients likely to respond to HER2-targeted therapies. The broad applicability of the assay was demonstrated by measuring HER2 expression profiles on multiple tumor types, and on normal and diseased heart tissues., (Copyright © 2013 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Receptor tyrosine kinases fall into distinct classes based on their inferred signaling networks.
- Author
-
Wagner JP, Wolf-Yadlin A, Sevecka M, Grenier JK, Root DE, Lauffenburger DA, and MacBeath G
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Enzymologic physiology, HEK293 Cells, Humans, Receptor Protein-Tyrosine Kinases genetics, Models, Biological, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction physiology
- Abstract
Although many anticancer drugs that target receptor tyrosine kinases (RTKs) provide clinical benefit, their long-term use is limited by resistance that is often attributed to increased abundance or activation of another RTK that compensates for the inhibited receptor. To uncover common and unique features in the signaling networks of RTKs, we measured time-dependent signaling in six isogenic cell lines, each expressing a different RTK as downstream proteins were systematically perturbed by RNA interference. Network models inferred from the data revealed a conserved set of signaling pathways and RTK-specific features that grouped the RTKs into three distinct classes: (i) an EGFR/FGFR1/c-Met class constituting epidermal growth factor receptor, fibroblast growth factor receptor 1, and the hepatocyte growth factor receptor c-Met; (ii) an IGF-1R/NTRK2 class constituting insulin-like growth factor 1 receptor and neurotrophic tyrosine receptor kinase 2; and (iii) a PDGFRβ class constituting platelet-derived growth factor receptor β. Analysis of cancer cell line data showed that many RTKs of the same class were coexpressed and that increased abundance of an RTK or its cognate ligand frequently correlated with resistance to a drug targeting another RTK of the same class. In contrast, abundance of an RTK or ligand of one class generally did not affect sensitivity to a drug targeting an RTK of a different class. Thus, classifying RTKs by their inferred networks and then therapeutically targeting multiple receptors within a class may delay or prevent the onset of resistance.
- Published
- 2013
- Full Text
- View/download PDF
38. Family-wide investigation of PDZ domain-mediated protein-protein interactions implicates β-catenin in maintaining the integrity of tight junctions.
- Author
-
Gujral TS, Karp ES, Chan M, Chang BH, and MacBeath G
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Carrier Proteins chemistry, Carrier Proteins metabolism, Cell Adhesion Molecules, Cell Adhesion Molecules, Neuronal chemistry, Cell Adhesion Molecules, Neuronal metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Membrane metabolism, Dogs, Fluorescence Polarization, Guanylate Kinases, HEK293 Cells, Humans, Madin Darby Canine Kidney Cells, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, PDZ Domains, Protein Interaction Domains and Motifs, RNA Interference, RNA, Small Interfering metabolism, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, beta Catenin antagonists & inhibitors, beta Catenin genetics, Tight Junctions metabolism, beta Catenin metabolism
- Abstract
β-catenin is a multifunctional protein that plays a critical role in cell-cell contacts and signal transduction. β-catenin has previously been shown to interact with PDZ-domain-containing proteins through its C terminus. Using protein microarrays comprising 206 mouse PDZ domains, we identified 26 PDZ-domain-mediated interactions with β-catenin and confirmed them biochemically and in cellular lysates. Many of the previously unreported interactions involved proteins with annotated roles in tight junctions. We found that four tight-junction-associated PDZ proteins-Scrib, Magi-1, Pard3, and ZO-3-colocalize with β-catenin at the plasma membrane. Disrupting these interactions by RNA interference, overexpression of PDZ domains, or overexpression of the β-catenin C terminus altered localization of the full-length proteins, weakened tight junctions, and decreased cellular adhesion. These results suggest that β-catenin serves as a scaffold to establish the location and function of tight-junction-associated proteins., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. Phosphotyrosine signaling proteins that drive oncogenesis tend to be highly interconnected.
- Author
-
Koytiger G, Kaushansky A, Gordus A, Rush J, Sorger PK, and MacBeath G
- Subjects
- HEK293 Cells, Humans, Oncogene Proteins metabolism, Protein Binding, Protein Interaction Domains and Motifs, Protein Interaction Maps, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction, Carcinogenesis metabolism, Intracellular Signaling Peptides and Proteins metabolism, Phosphotyrosine metabolism
- Abstract
Mutation and overexpression of receptor tyrosine kinases or the proteins they regulate serve as oncogenic drivers in diverse cancers. To better understand receptor tyrosine kinase signaling and its link to oncogenesis, we used protein microarrays to systematically and quantitatively measure interactions between virtually every SH2 or PTB domain encoded in the human genome and all known sites of tyrosine phosphorylation on 40 receptor tyrosine kinases and on most of the SH2 and PTB domain-containing adaptor proteins. We found that adaptor proteins, like RTKs, have many high affinity bindings sites for other adaptor proteins. In addition, proteins that drive cancer, including both receptors and adaptor proteins, tend to be much more highly interconnected via networks of SH2 and PTB domain-mediated interactions than nononcogenic proteins. Our results suggest that network topological properties such as connectivity can be used to prioritize new drug targets in this well-studied family of signaling proteins.
- Published
- 2013
- Full Text
- View/download PDF
40. Suppression of host p53 is critical for Plasmodium liver-stage infection.
- Author
-
Kaushansky A, Ye AS, Austin LS, Mikolajczak SA, Vaughan AM, Camargo N, Metzger PG, Douglass AN, MacBeath G, and Kappe SH
- Subjects
- Animals, Autophagy, Cell Proliferation, Cell Survival, Hepatocytes metabolism, Hepatocytes parasitology, Host-Parasite Interactions, Imidazoles pharmacology, Life Cycle Stages, Mice, Mice, Transgenic, Mutation, Piperazines pharmacology, Plasmodium yoelii growth & development, Plasmodium yoelii metabolism, Protein Array Analysis, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Liver parasitology, Plasmodium yoelii pathogenicity, Tumor Suppressor Protein p53 antagonists & inhibitors
- Abstract
Plasmodium parasites infect the liver and replicate inside hepatocytes before they invade erythrocytes and trigger clinical malaria. Analysis of host signaling pathways affected by liver-stage infection could provide critical insights into host-pathogen interactions and reveal targets for intervention. Using protein lysate microarrays, we found that Plasmodium yoelii rodent malaria parasites perturb hepatocyte regulatory pathways involved in cell survival, proliferation, and autophagy. Notably, the prodeath protein p53 was substantially decreased in infected hepatocytes, suggesting that it could be targeted by the parasite to foster survival. Indeed, mice that express increased levels of p53 showed reduced liver-stage parasite burden, whereas p53 knockout mice suffered increased liver-stage burden. Furthermore, boosting p53 levels with the use of the small molecule Nutlin-3 dramatically reduced liver-stage burden in vitro and in vivo. We conclude that perturbation of the hepatocyte p53 pathway critically impacts parasite survival. Thus, host pathways might constitute potential targets for host-based antimalarial prophylaxis., (Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
41. Cross-talk between receptor tyrosine kinase and tumor necrosis factor-α signaling networks regulates apoptosis but not proliferation.
- Author
-
Beyer EM and MacBeath G
- Subjects
- Anthracenes pharmacology, Cell Proliferation, HEK293 Cells, Humans, Imidazoles pharmacology, Intercellular Signaling Peptides and Proteins physiology, Mitogen-Activated Protein Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinases metabolism, Phenotype, Protein Interaction Maps, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins c-raf metabolism, Pyridines pharmacology, Tumor Necrosis Factor-alpha physiology, Apoptosis, Receptor Cross-Talk, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction, Tumor Necrosis Factor-alpha metabolism
- Abstract
Although many of the signaling networks activated by receptor tyrosine kinases (RTKs) and cytokine receptors are well understood, how these networks interconnect is much less clear. We set out to determine how cells respond to simultaneous exposure to opposing signals and how their downstream networks process this information. Using six isogenic cell lines, each stably transfected with a different RTK, we found that, in each case, the cognate growth factor induced proliferation, whereas TNFα induced apoptosis. Surprisingly, when the cells were treated simultaneously with growth factor and TNFα, the growth factor enhanced, rather than antagonized, TNFα-induced cell death. In contrast, TNFα had no effect on growth factor-induced proliferation, suggesting that cross-talk between these networks is unidirectional. A quantitative, system-wide study of signaling at early and late time points corroborated this observation: proteins in the RTK networks were not affected by TNFα treatment, but proteins in the TNFα network were affected by growth factors. These studies also highlighted the stress mitogen-activated protein kinase proteins p38 and c-Jun N-terminal kinase as the key nodes of signal integration, and their activation states at an early time point correlated well with subsequent measurements of apoptosis. Knocking down cRaf reduced the growth factor enhancement of TNFα-induced apoptosis, highlighting its role as a regulator of network cross-talk upstream of p38 and c-Jun N-terminal kinase. Overall, we found that when cells encounter conflicting stimuli, their phenotypic response is determined not by the sum of isolated processes, but by how their signaling networks interconnect. This underscores the need to build mechanistic models of network integration as a first step in predicting cellular behavior in complex settings and in rationally designing combination therapies.
- Published
- 2012
- Full Text
- View/download PDF
42. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks.
- Author
-
Lee MJ, Ye AS, Gardino AK, Heijink AM, Sorger PK, MacBeath G, and Yaffe MB
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Caspase 8, Cell Line, Tumor, DNA Damage, ErbB Receptors metabolism, Female, Humans, Metabolic Networks and Pathways, Models, Biological, Antineoplastic Agents administration & dosage, Apoptosis, Breast Neoplasms drug therapy, Drug Therapy, Combination methods, ErbB Receptors antagonists & inhibitors, Signal Transduction
- Abstract
Crosstalk and complexity within signaling pathways and their perturbation by oncogenes limit component-by-component approaches to understanding human disease. Network analysis of how normal and oncogenic signaling can be rewired by drugs may provide opportunities to target tumors with high specificity and efficacy. Using targeted inhibition of oncogenic signaling pathways, combined with DNA-damaging chemotherapy, we report that time-staggered EGFR inhibition, but not simultaneous coadministration, dramatically sensitizes a subset of triple-negative breast cancer cells to genotoxic drugs. Systems-level analysis-using high-density time-dependent measurements of signaling networks, gene expression profiles, and cell phenotypic responses in combination with mathematical modeling-revealed an approach for altering the intrinsic state of the cell through dynamic rewiring of oncogenic signaling pathways. This process converts these cells to a less tumorigenic state that is more susceptible to DNA damage-induced cell death by reactivation of an extrinsic apoptotic pathway whose function is suppressed in the oncogene-addicted state., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. A dual array-based approach to assess the abundance and posttranslational modification state of signaling proteins.
- Author
-
Luckert K, Gujral TS, Chan M, Sevecka M, Joos TO, Sorger PK, Macbeath G, and Pötz O
- Subjects
- Cells, Cultured, Culture Media, Humans, Protein Processing, Post-Translational, Proteins metabolism, Signal Transduction
- Abstract
A system-wide analysis of cell signaling requires detecting and quantifying many different proteins and their posttranslational modification states in the same cellular sample. Here, we present Protocols for two miniaturized, array-based methods, one of which provides detailed information on a central signaling protein and the other of which provides a broad characterization of the surrounding signaling network. We describe a bead-based array and its use in characterizing the different forms and functions of β-catenin, as well as lysate microarrays (reverse-phase protein arrays) and their use in detecting and quantifying proteins involved in the canonical and noncanonical Wnt signaling pathways. As an application of this dual approach, we characterized the state of β-catenin signaling in cell lysates and linked these molecule-specific data with pathway-wide changes in signaling. The Protocols described here provide detailed instructions for cell culture methods, bead arrays, and lysate microarrays and outline how to use these complementary approaches to obtain insight into a complex network at a systems level.
- Published
- 2012
- Full Text
- View/download PDF
44. A systematic family-wide investigation reveals that ~30% of mammalian PDZ domains engage in PDZ-PDZ interactions.
- Author
-
Chang BH, Gujral TS, Karp ES, BuKhalid R, Grantcharova VP, and MacBeath G
- Subjects
- Animals, Cell Line, Dimerization, Fluorescence Polarization, Humans, Mice, Protein Array Analysis, Protein Binding, Protein Interaction Maps, Proteins chemistry, Proteome chemistry, Proteome metabolism, PDZ Domains, Proteins metabolism
- Abstract
PDZ domains are independently folded modules that typically mediate protein-protein interactions by binding to the C termini of their target proteins. However, in a few instances, PDZ domains have been reported to dimerize with other PDZ domains. To investigate this noncanonical-binding mode further, we used protein microarrays comprising virtually every mouse PDZ domain to systematically query all possible PDZ-PDZ pairs. We then used fluorescence polarization to retest and quantify interactions and coaffinity purification to test biophysically validated interactions in the context of their full-length proteins. Overall, we discovered 37 PDZ-PDZ interactions involving 46 PDZ domains (~30% of all PDZ domains tested), revealing that dimerization is a more frequently used binding mode than was previously appreciated. This suggests that many PDZ domains evolved to form multiprotein complexes by simultaneously interacting with more than one ligand., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. Tensin2 is a novel mediator in thrombopoietin (TPO)-induced cellular proliferation by promoting Akt signaling.
- Author
-
Jung AS, Kaushansky A, Macbeath G, and Kaushansky K
- Subjects
- Genome, Human, Humans, Protein Array Analysis, Protein Interaction Domains and Motifs, Tensins, Cell Proliferation, Microfilament Proteins physiology, Phosphoric Monoester Hydrolases physiology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Thrombopoietin pharmacology
- Abstract
Thrombopoietin (TPO) and its receptor c-Mpl are essential in the regulation of the hematopoietic stem and progenitors cells as well as for the differentiation of megakaryocytes into mature platelets. Once TPO binds to its receptor, an intracellular signaling process is initiated through Janus kinase (JAK-2)-induced phosphorylation of the c-Mpl intracellular domain. Although some protein mediators that transmit the effects of TPO have been identified, many remain undiscovered. Using an unbiased approach with peptide microarrays that contained virtually every Src Homology (SH)2 and Phosphotyrosine Binding (PTB) domains in the human genome, we discovered a previously unreported interaction between c-Mpl at phospho-Tyrosine631 (pY 631) and Tensin2, a protein for which limited information is available. Confirming the findings of the microarrays, we discovered that Tensin2 co-precipitates with a pY 631 bearing peptide. Furthermore, we found that Tensin2 becomes phosphorylated in a TPO dependent manner. The functional consequence of Tensin2 was tested via knockdown of Tensin2, which dramatically decreased TPO-dependent cellular proliferation of UT7-TPO cell line as well as their activation of Akt signaling. These studies affirm the use of these arrays as an unbiased screening tool of protein-protein interactions. We conclude that Tensin2 is an important new mediator in TPO/c-Mpl pathway and has a positive affect on cellular growth, at least in part through its effect on the PI3K/Akt signaling.
- Published
- 2011
- Full Text
- View/download PDF
46. Lysate microarrays enable high-throughput, quantitative investigations of cellular signaling.
- Author
-
Sevecka M, Wolf-Yadlin A, and MacBeath G
- Subjects
- Antigens immunology, Biomarkers metabolism, Cell Line, Cross Reactions, Humans, Intracellular Signaling Peptides and Proteins immunology, Intracellular Signaling Peptides and Proteins metabolism, Protein Processing, Post-Translational, Antibodies metabolism, Antigens metabolism, Protein Array Analysis methods, Signal Transduction
- Abstract
Lysate microarrays (reverse-phase protein arrays) hold great promise as a tool for systems-level investigations of signaling and multiplexed analyses of disease biomarkers. To date, however, widespread use of this technology has been limited by questions concerning data quality and the specificity of detection reagents. To address these concerns, we developed a strategy to identify high-quality reagents for use with lysate microarrays. In total, we tested 383 antibodies for their ability to quantify changes in protein abundance or modification in 20 biological contexts across 17 cell lines. Antibodies yielding significant differences in signal were further evaluated by immunoblotting and 82 passed our rigorous criteria. The large-scale data set from our screen revealed that cell fate decisions are encoded not just by the identities of proteins that are activated, but by differences in their signaling dynamics as well. Overall, our list of validated antibodies and associated protocols establish lysate microarrays as a robust tool for systems biology.
- Published
- 2011
- Full Text
- View/download PDF
47. High- and low-affinity epidermal growth factor receptor-ligand interactions activate distinct signaling pathways.
- Author
-
Krall JA, Beyer EM, and MacBeath G
- Subjects
- Cell Line, Humans, Ligands, Phospholipase C gamma metabolism, Protein Binding, STAT Transcription Factors metabolism, ErbB Receptors metabolism, Signal Transduction
- Abstract
Signaling mediated by the Epidermal Growth Factor Receptor (EGFR) is crucial in normal development, and aberrant EGFR signaling has been implicated in a wide variety of cancers. Here we find that the high- and low-affinity interactions between EGFR and its ligands activate different signaling pathways. While high-affinity ligand binding is sufficient for activation of most canonical signaling pathways, low-affinity binding is required for the activation of the Signal transducers and activators of transcription (Stats) and Phospholipase C-gamma 1 (PLCγ1). As the Stat proteins are involved in many cellular responses including proliferation, migration and apoptosis, these results assign a function to low-affinity interactions that has been omitted from computational models of EGFR signaling. The existence of receptors with distinct signaling properties provides a way for EGFR to respond to different concentrations of the same ligand in qualitatively different ways.
- Published
- 2011
- Full Text
- View/download PDF
48. Detecting and quantifying multiple proteins in clinical samples in high-throughput using antibody microarrays.
- Author
-
Knickerbocker T and MacBeath G
- Subjects
- Animals, Biomarkers analysis, Biomarkers blood, Humans, Image Processing, Computer-Assisted, Immunoassay instrumentation, Printing, Protein Array Analysis instrumentation, Proteins immunology, Time Factors, Antibodies, Immobilized immunology, Immunoassay methods, Protein Array Analysis methods, Proteins analysis
- Abstract
Many diagnostic and prognostic tests performed in the clinic today rely on the sensitive detection and quantification of a single protein, usually by means of an immunoassay. Even in the case of monogenic diseases, however, single markers are often insufficient to provide highly reliable predictions of disease onset, and the accuracy of these predictions only decreases for polygenic diseases and for very early detection or prediction. Recent studies have shown that predictive reliability increases dramatically when multiple markers are analyzed simultaneously. Antibody microarrays provide a powerful way to quantify the abundance of many different proteins simultaneously in a variety of sample types, including serum, urine, and tissue explants. Because the assay is highly miniaturized, very little sample is required and the assay can be performed in high-throughput. Using antibody microarrays, we have been able to identify prognostic markers of early mortality in patients with end-stage renal disease and have built multivariate models based on these markers. We anticipate that antibody microarrays will prove similarly useful in other discovery-based efforts and may ultimately enjoy routine use in clinical labs.
- Published
- 2011
- Full Text
- View/download PDF
49. Tyrosine-phosphorylated caveolin-1 blocks bacterial uptake by inducing Vav2-RhoA-mediated cytoskeletal rearrangements.
- Author
-
Boettcher JP, Kirchner M, Churin Y, Kaushansky A, Pompaiah M, Thorn H, Brinkmann V, Macbeath G, and Meyer TF
- Subjects
- Caveolin 1 genetics, Caveolin 1 pharmacology, Cell Line, Tumor, Fimbriae, Bacterial metabolism, Host-Pathogen Interactions, Humans, Neisseria gonorrhoeae physiology, Phosphorylation, Proto-Oncogene Proteins c-vav genetics, Proto-Oncogene Proteins c-vav metabolism, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Caveolin 1 metabolism, Cytoskeleton metabolism, Epithelial Cells microbiology, Gene Expression Regulation, Neisseria gonorrhoeae pathogenicity, Signal Transduction, Tyrosine metabolism
- Abstract
Certain bacterial adhesins appear to promote a pathogen's extracellular lifestyle rather than its entry into host cells. However, little is known about the stimuli elicited upon such pathogen host-cell interactions. Here, we report that type IV pili (Tfp)-producing Neisseria gonorrhoeae (P(+)GC) induces an immediate recruitment of caveolin-1 (Cav1) in the host cell, which subsequently prevents bacterial internalization by triggering cytoskeletal rearrangements via downstream phosphotyrosine signaling. A broad and unbiased analysis of potential interaction partners for tyrosine-phosphorylated Cav1 revealed a direct interaction with the Rho-family guanine nucleotide exchange factor Vav2. Both Vav2 and its substrate, the small GTPase RhoA, were found to play a direct role in the Cav1-mediated prevention of bacterial uptake. Our findings, which have been extended to enteropathogenic Escherichia coli, highlight how Tfp-producing bacteria avoid host cell uptake. Further, our data establish a mechanistic link between Cav1 phosphorylation and pathogen-induced cytoskeleton reorganization and advance our understanding of caveolin function., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2010
- Full Text
- View/download PDF
50. Tarp regulates early Chlamydia-induced host cell survival through interactions with the human adaptor protein SHC1.
- Author
-
Mehlitz A, Banhart S, Mäurer AP, Kaushansky A, Gordus AG, Zielecki J, Macbeath G, and Meyer TF
- Subjects
- Bacterial Proteins genetics, Cell Survival, Chlamydia Infections genetics, Chlamydia trachomatis genetics, Gene Expression Regulation, HeLa Cells, Humans, Protein Structure, Tertiary, Shc Signaling Adaptor Proteins genetics, Src Homology 2 Domain-Containing, Transforming Protein 1, Apoptosis, Bacterial Proteins metabolism, Chlamydia Infections metabolism, Chlamydia trachomatis metabolism, Shc Signaling Adaptor Proteins metabolism, Signal Transduction
- Abstract
Many bacterial pathogens translocate effector proteins into host cells to manipulate host cell functions. Here, we used a protein microarray comprising virtually all human SRC homology 2 (SH2) and phosphotyrosine binding domains to comprehensively and quantitatively assess interactions between host cell proteins and the early phase Chlamydia trachomatis effector protein translocated actin-recruiting phosphoprotein (Tarp), which is rapidly tyrosine phosphorylated upon host cell entry. We discovered numerous novel interactions between human SH2 domains and phosphopeptides derived from Tarp. The adaptor protein SHC1 was among Tarp's strongest interaction partners. Transcriptome analysis of SHC1-dependent gene regulation during infection indicated that SHC1 regulates apoptosis- and growth-related genes. SHC1 knockdown sensitized infected host cells to tumor necrosis factor-induced apoptosis. Collectively, our findings reveal a critical role for SHC1 in early C. trachomatis-induced cell survival and suggest that Tarp functions as a multivalent phosphorylation-dependent signaling hub that is important during the early phase of chlamydial infection.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.