Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oncologist ; 27(10): 864-873, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35948003

ABSTRACT

BACKGROUND: The safety and efficacy of brentuximab vedotin (BV), an antibody-drug conjugate directed to the CD30 antigen, has been assessed in several trials in patients with peripheral T-cell lymphoma (PTCL), cutaneous T-cell lymphoma (CTCL), or B-cell non-Hodgkin lymphoma (NHL). The objective of this research was to examine the relationship between CD30 expression level and clinical response to BV. PATIENTS AND METHODS: We analyzed response in patients treated with BV monotherapy in 5 prospective clinical studies in relapsed or refractory PTCL, CTCL, or B-cell NHL. CD30 expression was assessed by immunohistochemistry (IHC) using the Ber H2 antibody for 275 patients. RESULTS: Across all 5 studies, 140 (50.9%) patients had tumors with CD30 expression <10%, including 60 (21.8%) with undetectable CD30 by IHC. No significant differences were observed for any study in overall response rates between patients with CD30 expression ≥10% or <10%. Median duration of response was also similar in the CD30 ≥10% and <10% groups for all studies. CONCLUSIONS: In this analysis of studies across a range of CD30-expressing lymphomas, CD30 expression alone, as measured by standard IHC, does not predict clinical benefit from BV, making the determination of a threshold level of expression uncertain.


Subject(s)
Immunoconjugates , Lymphoma, T-Cell, Peripheral , Brentuximab Vedotin , Humans , Immunoconjugates/adverse effects , Ki-1 Antigen/metabolism , Lymphoma, T-Cell, Peripheral/drug therapy , Prospective Studies
2.
Eur J Cancer ; 148: 411-421, 2021 05.
Article in English | MEDLINE | ID: mdl-33794441

ABSTRACT

INTRODUCTION: Mycosis fungoides (MF), the most common type of cutaneous T-cell lymphoma, can lead to disfiguring lesions, debilitating pruritus and frequent skin infections. This study assessed response to brentuximab vedotin in patients with MF in the phase III ALCANZA study. METHODS: Baseline CD30 levels and large-cell transformation (LCT) status were centrally reviewed in patients with previously-treated CD30-positive MF using ≥2 skin biopsies obtained at screening; eligible patients required ≥1 biopsy with ≥10% CD30 expression. Patients were categorised as CD30min < 10% (≥1 biopsy with <10% CD30 expression), or CD30min ≥ 10% (all biopsies with ≥10% CD30 expression) and baseline LCT present or absent. Efficacy analyses were the proportion of patients with objective response lasting ≥4 months (ORR4) and progression-free survival (PFS). RESULTS: Clinical activity with brentuximab vedotin was observed across all CD30 expression levels in patients with ≥1 biopsy showing ≥10% CD30 expression. Superior ORR4 was observed with brentuximab vedotin versus physician's choice in patients: with CD30min < 10% (40.9% versus 9.5%), with CD30min ≥ 10% (57.1% versus 10.3%), with LCT (64.7% versus 17.6%) and without LCT (38.7% versus 6.5%). Brentuximab vedotin improved median PFS versus physician's choice in patients: with CD30min < 10% (16.7 versus 2.3 months), with CD30min ≥ 10% (15.5 versus 3.9 months), with LCT (15.5 versus 2.8 months) and without LCT (16.1 versus 3.5 months). Safety profiles were generally comparable across subgroups. CONCLUSION: These exploratory analyses demonstrated that brentuximab vedotin improved rates of ORR4 and PFS versus physician's choice in patients with CD30-positive MF and ≥1 biopsy showing ≥10% CD30 expression, regardless of LCT status. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov, NCT01578499.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Brentuximab Vedotin/therapeutic use , Choice Behavior , Decision Support Techniques , Ki-1 Antigen/metabolism , Mycosis Fungoides/drug therapy , Physicians/statistics & numerical data , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , International Agencies , Male , Middle Aged , Mycosis Fungoides/metabolism , Mycosis Fungoides/pathology , Physicians/psychology , Prognosis , Retrospective Studies , Survival Rate , Young Adult
3.
PLoS Comput Biol ; 12(4): e1004827, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27035903

ABSTRACT

Understanding the molecular pathways by which oncogenes drive cancerous cell growth, and how dependence on such pathways varies between tumors could be highly valuable for the design of anti-cancer treatment strategies. In this work we study how dependence upon the canonical PI3K and MAPK cascades varies across HER2+ cancers, and define biomarkers predictive of pathway dependencies. A panel of 18 HER2+ (ERBB2-amplified) cell lines representing a variety of indications was used to characterize the functional and molecular diversity within this oncogene-defined cancer. PI3K and MAPK-pathway dependencies were quantified by measuring in vitro cell growth responses to combinations of AKT (MK2206) and MEK (GSK1120212; trametinib) inhibitors, in the presence and absence of the ERBB3 ligand heregulin (NRG1). A combination of three protein measurements comprising the receptors EGFR, ERBB3 (HER3), and the cyclin-dependent kinase inhibitor p27 (CDKN1B) was found to accurately predict dependence on PI3K/AKT vs. MAPK/ERK signaling axes. Notably, this multivariate classifier outperformed the more intuitive and clinically employed metrics, such as expression of phospho-AKT and phospho-ERK, and PI3K pathway mutations (PIK3CA, PTEN, and PIK3R1). In both cell lines and primary patient samples, we observed consistent expression patterns of these biomarkers varies by cancer indication, such that ERBB3 and CDKN1B expression are relatively high in breast tumors while EGFR expression is relatively high in other indications. The predictability of the three protein biomarkers for differentiating PI3K/AKT vs. MAPK dependence in HER2+ cancers was confirmed using external datasets (Project Achilles and GDSC), again out-performing clinically used genetic markers. Measurement of this minimal set of three protein biomarkers could thus inform treatment, and predict mechanisms of drug resistance in HER2+ cancers. More generally, our results show a single oncogenic transformation can have differing effects on cell signaling and growth, contingent upon the molecular and cellular context.


Subject(s)
MAP Kinase Signaling System , Neoplasms/genetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Knockdown Techniques , Genes, erbB-2 , Humans , MAP Kinase Signaling System/genetics , Mutation , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
4.
Am J Pathol ; 183(5): 1446-1460, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035511

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.


Subject(s)
Genetic Heterogeneity , Neoplasms/classification , Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Single-Cell Analysis/methods , 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
5.
Sci Signal ; 6(288): ra68, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23943608

ABSTRACT

Crosstalk and compensatory circuits within cancer signaling networks limit the activity of most targeted therapies. For example, altered signaling in the networks activated by the ErbB family of receptors, particularly in ERBB2-amplified cancers, contributes to drug resistance. We developed a multiscale systems model of signaling networks in ERBB2-amplified breast cancer to quantitatively investigate relationships between biomarkers (markers of network activity) and combination drug efficacy. This model linked ErbB receptor family signaling to breast tumor growth through two kinase cascades: the PI3K/AKT survival pathway and the Ras/MEK/ERK growth and proliferation pathway. The model predicted molecular mechanisms of resistance to individual therapeutics. In particular, ERBB2-amplified breast cancer cells stimulated with the ErbB3 ligand heregulin were resistant to growth arrest induced by inhibitors of AKT and MEK or coapplication of two inhibitors of the receptor ErbB2 [Herceptin (trastuzumab) and Tykerb (lapatinib)]. We used model simulations to predict the response of ErbB2-positive breast cancer xenografts to combination therapies and verified these predictions in mice. Treatment with trastuzumab, lapatinib, and the ErbB3 inhibitor MM-111 was more effective in inhibiting tumor growth than the combination of AKT and MEK inhibitors and even induced tumor regression, indicating that targeting both ErbB3 and ErbB2 may be an improved therapeutic approach for ErbB2-positive breast cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Models, Biological , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction/physiology , Animals , Antibodies, Bispecific , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/metabolism , Breast Neoplasms/physiopathology , Computer Simulation , Feedback, Physiological/physiology , Female , Lapatinib , MAP Kinase Signaling System/drug effects , Mice , Neuregulin-1 , Oncogene Protein v-akt/antagonists & inhibitors , Quinazolines , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Trastuzumab
6.
Cancer Res ; 70(6): 2485-94, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215504

ABSTRACT

ErbB3 is a critical activator of phosphoinositide 3-kinase (PI3K) signaling in epidermal growth factor receptor (EGFR; ErbB1), ErbB2 [human epidermal growth factor receptor 2 (HER2)], and [hepatocyte growth factor receptor (MET)] addicted cancers, and reactivation of ErbB3 is a prominent method for cancers to become resistant to ErbB inhibitors. In this study, we evaluated the in vivo efficacy of a therapeutic anti-ErbB3 antibody, MM-121. We found that MM-121 effectively blocked ligand-dependent activation of ErbB3 induced by either EGFR, HER2, or MET. Assessment of several cancer cell lines revealed that MM-121 reduced basal ErbB3 phosphorylation most effectively in cancers possessing ligand-dependent activation of ErbB3. In those cancers, MM-121 treatment led to decreased ErbB3 phosphorylation and, in some instances, decreased ErbB3 expression. The efficacy of single-agent MM-121 was also examined in xenograft models. A machine learning algorithm found that MM-121 was most effective against xenografts with evidence of ligand-dependent activation of ErbB3. We subsequently investigated whether MM-121 treatment could abrogate resistance to anti-EGFR therapies by preventing reactivation of ErbB3. We observed that an EGFR mutant lung cancer cell line (HCC827), made resistant to gefitinib by exogenous heregulin, was resensitized by MM-121. In addition, we found that a de novo lung cancer mouse model induced by EGFR T790M-L858R rapidly became resistant to cetuximab. Resistance was associated with an increase in heregulin expression and ErbB3 activation. However, concomitant cetuximab treatment with MM-121 blocked reactivation of ErbB3 and resulted in a sustained and durable response. Thus, these results suggest that targeting ErbB3 with MM-121 can be an effective therapeutic strategy for cancers with ligand-dependent activation of ErbB3.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neoplasms/therapy , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , CHO Cells , Cell Line, Tumor , Cetuximab , Cricetinae , Cricetulus , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Ligands , Mice , Neoplasms/metabolism , Neuregulin-1/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-3/metabolism , Xenograft Model Antitumor Assays
7.
Curr Opin Cell Biol ; 21(1): 74-81, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19167872

ABSTRACT

Theorists have long speculated on the mechanisms driving directed and spontaneous cell polarization. Recently, experimentalists have uncovered many of the mechanisms underlying polarization, enabling these models to be directly tested. In the process, they have demonstrated the explanatory and predictive value of these models and, at the same time, uncovered additional complexities not currently explained by them. In this review, we discuss some of main theories regarding cell polarization and highlight how the intersection of mathematical and experimental biology has yielded new insights into these mechanisms in the case of budding yeast and eukaryotic chemotaxis.


Subject(s)
Cell Polarity , Models, Biological , Animals , Eukaryotic Cells/metabolism , Saccharomyces cerevisiae/cytology
8.
PLoS Comput Biol ; 3(3): e36, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17367201

ABSTRACT

Directed cell migration in response to chemical cues, also known as chemotaxis, is an important physiological process involved in wound healing, foraging, and the immune response. Cell migration requires the simultaneous formation of actin polymers at the leading edge and actomyosin complexes at the sides and back of the cell. An unresolved question in eukaryotic chemotaxis is how the same chemoattractant signal determines both the cell's front and back. Recent experimental studies have begun to reveal the biochemical mechanisms necessary for this polarized cellular response. We propose a mathematical model of neutrophil gradient sensing and polarization based on experimentally characterized biochemical mechanisms. The model demonstrates that the known dynamics for Rho GTPase and phosphatidylinositol-3-kinase (PI3K) activation are sufficient for both gradient sensing and polarization. In particular, the model demonstrates that these mechanisms can correctly localize the "front" and "rear" pathways in response to both uniform concentrations and gradients of chemical attractants, including in actin-inhibited cells. Furthermore, the model predictions are robust to the values of many parameters. A key result of the model is the proposed coincidence circuit involving PI3K and Ras that obviates the need for the "global inhibitors" proposed, though never experimentally verified, in many previous mathematical models of eukaryotic chemotaxis. Finally, experiments are proposed to (in)validate this model and further our understanding of neutrophil chemotaxis.


Subject(s)
Chemotactic Factors/administration & dosage , Chemotaxis/physiology , Models, Biological , Neutrophils/cytology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Cell Polarity/drug effects , Cell Polarity/physiology , Chemotaxis/drug effects , Computer Simulation
9.
Phys Biol ; 3(3): 190-9, 2006 Sep 25.
Article in English | MEDLINE | ID: mdl-17021383

ABSTRACT

A key mediator of eukaryotic chemotaxis is the asymmetric accumulation of phosphatidylinositol-3,4,5-triphosphate (PIP3) on the cell membrane. Recent work has focused on understanding how a shallow external gradient of chemoattractant leads to an amplified internal gradient of PIP3. In this paper we dissect what fraction of this amplification is derived biochemically by the signal transduction network and how much arises entirely from the effects of cell morphology. Here we identify and formalize the role of morphology in signal detection and demonstrate its effects through simulation and experiments. Our key result is that an asymmetric distribution of membrane accounts for approximately one-half of the measured amplification from ligand concentration to PIP3 production. We also show that the underlying biochemical network behaves as a linear amplifier in the micropipette assay.


Subject(s)
Cell Shape , Chemotaxis, Leukocyte , Phosphatidylinositol Phosphates/metabolism , Signal Transduction , Cell Membrane/chemistry , HL-60 Cells , Humans , Metabolic Networks and Pathways , Phosphatidylinositol Phosphates/analysis
10.
Proc Natl Acad Sci U S A ; 102(32): 11151-6, 2005 Aug 09.
Article in English | MEDLINE | ID: mdl-16076952

ABSTRACT

We present a numerical method for computing diffusive transport on a surface derived from image data. Our underlying discretization method uses a Cartesian grid embedded boundary method for computing the volume transport in a region consisting of all points a small distance from the surface. We obtain a representation of this region from image data by using a front propagation computation based on level set methods for solving the Hamilton-Jacobi and eikonal equations. We demonstrate that the method is second-order accurate in space and time and is capable of computing solutions on complex surface geometries obtained from image data of cells.


Subject(s)
Models, Biological , Systems Biology , Biological Transport/physiology , Diffusion , Mathematics , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...