Network


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

Hotspot


Dive into the research topics where Chelsea J. Gudgeon is active.

Publication


Featured researches published by Chelsea J. Gudgeon.


Blood | 2014

Cellular determinants for preclinical activity of a novel CD33/CD3 bispecific T-cell engager (BiTE) antibody, AMG 330, against human AML

George S. Laszlo; Chelsea J. Gudgeon; Kimberly H. Harrington; Justine Dell’Aringa; Kathryn J. Newhall; Gary D. Means; Angus M. Sinclair; Roman Kischel; Stanley R. Frankel; Roland B. Walter

CD33 is a valid target for acute myeloid leukemia (AML) but has proven challenging for antibody-drug conjugates. Herein, we investigated the cellular determinants for the activity of the novel CD33/CD3-directed bispecific T-cell engager antibody, AMG 330. In the presence of T cells, AMG 330 was highly active against human AML cell lines and primary AML cells in a dose- and effector to target cell ratio-dependent manner. Using cell lines engineered to express wild-type CD33 at increased levels, we found a quantitative relationship between AMG 330 cytotoxicity and CD33 expression; in contrast, AMG 330 cytotoxicity was neither affected by common CD33 single nucleotide polymorphisms nor expression of the adenosine triphosphate-binding cassette (ABC) transporter proteins, P-glycoprotein or breast cancer resistance protein. Unlike bivalent CD33 antibodies, AMG 330 did not reduce surface CD33 expression. The epigenetic modifier drugs, panobinostat and azacitidine, increased CD33 expression in some cell lines and augmented AMG 330-induced cytotoxicity. These findings demonstrate that AMG 330 has potent CD33-dependent cytolytic activity in vitro, which can be further enhanced with other clinically available therapeutics. As it neither modulates CD33 expression nor is affected by ABC transporter activity, AMG 330 is highly promising for clinical exploration as it may overcome some limitations of previous CD33-targeted agents.


PLOS ONE | 2015

The Broad Anti-AML Activity of the CD33/CD3 BiTE Antibody Construct, AMG 330, Is Impacted by Disease Stage and Risk

Kimberly H. Harrington; Chelsea J. Gudgeon; George S. Laszlo; Kathryn J. Newhall; Angus M. Sinclair; Stanley R. Frankel; Roman Kischel; Guang Chen; Roland B. Walter

The CD33/CD3-bispecific T-cell engaging (BiTE) antibody construct, AMG 330, potently lyses CD33+ leukemic cells in vitro. Using specimens from 41 patients with acute myeloid leukemia (AML), we studied the factors that might contribute to clinical response or resistance. For this purpose, thawed aliquots of primary AML samples were immunophenotypically characterized and subjected to various doses of AMG 330 in the presence or absence of healthy donor T-cells. After 48 hours, drug-specific cytotoxicity was quantified and correlated with CD33 expression levels, amounts of T-cells present, and other disease characteristics. AMG 330 caused modest cytotoxicity that was correlated with the amount of autologous T-cells (P = 0.0001) but not CD33 expression, as AMG 330 exerted marked cytotoxic effects in several specimens with minimal CD33 expression. With healthy donor T-cells added, AMG 330 cytotoxicity depended on the drug dose and effector:target (E:T) cell ratio. High cytotoxic activity was observed even with minimal CD33 expression, and AMG 330 cytotoxicity and CD33 expression correlated only at high E:T cell ratio and high AMG 330 doses (P<0.003). AMG 330 resulted in significantly higher cytotoxicity in specimens from patients with newly diagnosed AML than those with relapsed/refractory disease despite similar levels of CD33 on myeloblasts. AMG 330 cytotoxicity also appeared greater in specimens from patients with favorable-risk disease as compared to other specimens. Together, our data demonstrate that AMG 330 is highly active in primary AML specimens across the entire disease spectrum, while suggesting the presence of yet undefined, CD33-independent, relative resistance mechanisms in specific patient subsets.


Blood Cancer Journal | 2015

T-cell ligands modulate the cytolytic activity of the CD33/CD3 BiTE antibody construct, AMG 330.

George S. Laszlo; Chelsea J. Gudgeon; Kimberly H. Harrington; Roland B. Walter

Preclinical and emerging clinical studies demonstrate that bispecific T-cell engaging (BiTE) antibody constructs can potently lyse targeted tumor cells, but the determinants for their activity remain incompletely understood. Using human acute myeloid leukemia (AML) cell lines engineered to overexpress individual T-cell ligands, we found that expression of the inhibitory ligands, PD-L1 and PD-L2, reduced the cytolytic activity of the BiTE antibody construct targeting CD33, AMG 330; conversely, expression of the activating ligands, CD80 and CD86, augmented the cytotoxic activity of AMG 330. Consistent with these findings, treatment with an activating antibody directed at the co-stimulatory T-cell receptor, CD28, significantly increased AMG 330-induced cytotoxicity in human AML cell lines. Using specimens from 12 patients with newly diagnosed or relapsed/refractory AML, we found that activation of CD28 also increased the activity of AMG 330 in primary human AML cells (P=0.023). Together, our findings indicate that T-cell ligands and co-receptors modulate the anti-tumor activity of the CD33/CD3 BiTE antibody construct, AMG 330. These findings suggest that such ligands/co-receptors could serve as biomarkers of response and that co-treatment strategies with pharmacological modulators of T-cell receptor signaling could be utilized to further enhance the activity of this targeted therapeutic.


Clinical Cancer Research | 2016

Characterization of CD33/CD3 Tetravalent Bispecific Tandem Diabodies (TandAbs) for the Treatment of Acute Myeloid Leukemia

Uwe Reusch; Kimberly H. Harrington; Chelsea J. Gudgeon; Ivica Fucek; Kristina Ellwanger; Michael Weichel; Stefan Knackmuss; Eugene A. Zhukovsky; Judith A. Fox; Lori Kunkel; Jeanmarie Guenot; Roland B. Walter

Purpose: Randomized studies with gemtuzumab ozogamicin have validated CD33 as a target for antigen-specific immunotherapy of acute myelogenous leukemia (AML). Here, we investigated the potential of CD33/CD3-directed tandem diabodies (TandAbs) as novel treatment approach for AML. These tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a bivalent antibody and have a molecular weight exceeding the renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs. Experimental Design: We constructed a series of TandAbs composed of anti-CD33 and anti-CD3 variable domains of diverse binding affinities and profiled their functional properties in CD33+ human leukemia cell lines, xenograft models, and AML patient samples. Results: Our studies demonstrated that several CD33/CD3 TandAbs could induce potent, dose-dependent cytolysis of CD33+ AML cell lines. This effect was modulated by the effector-to-target cell ratio and strictly required the presence of T cells. Activation and proliferation of T cells and maximal AML cell cytolysis correlated with high avidity to both CD33 and CD3. High-avidity TandAbs were broadly active in primary specimens from patients with newly diagnosed or relapsed/refractory AML in vitro, with cytotoxic properties independent of CD33 receptor density and cytogenetic risk. Tumor growth delay and inhibition were observed in both prophylactic and established HL-60 xenograft models in immunodeficient mice. Conclusions: Our data show high efficacy of CD33/CD3 TandAbs in various preclinical models of human AML. Together, these findings support further study of CD33/CD3 TandAbs as novel immunotherapeutics for patients with AML. Clin Cancer Res; 22(23); 5829–38. ©2016 AACR.


PLOS ONE | 2013

AKT Signaling as a Novel Factor Associated with In Vitro Resistance of Human AML to Gemtuzumab Ozogamicin

David B. Rosen; Kimberly H. Harrington; James Cordeiro; Ling Y. Leung; Santosh Putta; Norman J. Lacayo; George S. Laszlo; Chelsea J. Gudgeon; Donna E. Hogge; Rachael E. Hawtin; Alessandra Cesano; Roland B. Walter

Gemtuzumab ozogamicin (GO), an immunoconjugate between an anti-CD33 antibody and a calicheamicin-γ1 derivative, induces remissions and improves survival in a subset of patients with acute myeloid leukemia (AML). As the mechanisms underlying GO and calicheamicin-γ1 resistance are incompletely understood, we herein used flow cytometry-based single cell network profiling (SCNP) assays to study cellular responses of primary human AML cells to GO. Our data indicate that the extent of DNA damage is quantitatively impacted by CD33 expression and drug efflux activity. However, although DNA damage is required for GO-induced cytotoxicity, it is not sufficient for effective cell kill, suggesting that downstream anti-apoptotic pathways may function as relevant resistance mechanisms. Supporting this notion, we found activated PI3K/AKT signaling to be associated with GO resistance in vitro in primary AML cells. Consistently, the investigational AKT inhibitor MK-2206 significantly sensitized various human AML cells to GO or free calicheamicin-γ1 with particularly pronounced effects in otherwise GO or free calicheamicin-γ1 -resistant cells. Likewise, MK-2206 also sensitized primary AML cells to calicheamicin-γ1. Together, our findings illustrate the capacity of SCNP assays to discover chemotherapy-related biological pathways and signaling networks relevant to GO-induced genotoxic stress. The identification of AKT signaling as being associated with GO resistance in vitro may provide a novel approach to improve the in vivo efficacy of GO/calicheamicin-γ1 and, by extrapolation, other DNA damage-based therapeutics.


Oncotarget | 2016

Expression and functional characterization of CD33 transcript variants in human acute myeloid leukemia

George S. Laszlo; Kimberly H. Harrington; Chelsea J. Gudgeon; Mary E. Beddoe; Matthew Fitzgibbon; Rhonda E. Ries; Jatinder K. Lamba; Martin W. McIntosh; Soheil Meshinchi; Roland B. Walter

With the demonstration of improved survival of some acute myeloid leukemia (AML) patients with the CD33 antibody-drug conjugate, gemtuzumab ozogamicin (GO), CD33 has been validated as a target for antigen-specific immunotherapy. Since previous studies identified a CD33 splice variant missing exon 2 (CD33∆E2) and, consequently, the immune-dominant membrane-distal V-set domain, we investigated the expression and functional characteristics of CD33 transcript variants in AML. In primary AML specimens, we not only found full-length CD33 (CD33FL) and CD33∆E2 but also corresponding variants containing an alternate exon 7 predicted to encode a CD33 protein lacking most of the intracellular domain (CD33E7a and, not previously described, CD33∆E2,E7a) in almost all cases. In acute leukemia cell sublines engineered to express individual CD33 splice variants, all splice variants had endocytic properties. CD33FL and CD33E7a mediated similar degrees of GO cytotoxicity, whereas CD33∆E2 and CD33∆E2,E7a could not serve as target for GO. Co-expression of CD33∆E2 did not interfere with CD33FL endocytosis and did not impact CD33FL-mediated GO cytotoxicity. Together, our findings document a greater-than-previously thought complexity of CD33 expression in human AML. They identify CD33 variants that lack exon 2 and are not recognized by current CD33-directed therapeutics as potential target for future unconjugated or conjugated antibodies.


Leukemia | 2014

Heterogeneity of Clonal Expansion and Maturation-Linked Mutation Acquisition in Hematopoietic Progenitors in Human Acute Myeloid Leukemia

Roland B. Walter; George S. Laszlo; Jack M. Lionberger; Jessica A. Pollard; Kimberly H. Harrington; Chelsea J. Gudgeon; Megan Othus; Shahin Rafii; Soheil Meshinchi; Frederick R. Appelbaum; Irwin D. Bernstein

Recent technological advances led to an appreciation of the genetic complexity of human acute myeloid leukemia (AML), but underlying progenitor cells remain poorly understood because their rarity precludes direct study. We developed a co-culture method integrating hypoxia, aryl hydrocarbon receptor inhibition and micro-environmental support via human endothelial cells to isolate these cells. X-chromosome inactivation studies of the least mature precursors derived following prolonged culture of CD34+/CD33− cells revealed polyclonal growth in highly curable AMLs, suggesting that mutations necessary for clonal expansion were acquired in more mature progenitors. Consistently, in core-binding factor (CBF) leukemias with known complementing mutations, immature precursors derived following prolonged culture of CD34+/CD33− cells harbored neither mutation or the CBF mutation alone, whereas more mature precursors often carried both mutations. These results were in contrast to those with leukemias with poor prognosis that showed clonal dominance in the least mature precursors. These data indicate heterogeneity among progenitors in human AML that may have prognostic and therapeutic implications.


American Journal of Hematology | 2013

Significance of Expression of ITGA5 and its Splice Variants in Acute Myeloid Leukemia: A Report from the Children’s Oncology Group

Roland B. Walter; George S. Laszlo; Todd A. Alonzo; Robert B. Gerbing; Shawn Levy; Matthew Fitzgibbon; Chelsea J. Gudgeon; Rhonda E. Ries; Kimberly H. Harrington; Susana C. Raimondi; Betsy Hirsch; Alan S. Gamis; Martin W. McIntosh; Soheil Meshinchi

Acute myeloid leukemia (AML) encompasses a heterogeneous group of diseases, and novel biomarkers for risk refinement and stratification are needed to optimize patient care. To identify novel risk factors, we performed transcriptome sequencing on 68 diagnostic AML samples and identified 2 transcript variants (–E2 and –E2/3) of the α‐subunit (ITGA5) of the very late antigen‐5 integrin. We then quantified expression of ITGA5 and these splice variants in specimens from participants of the AAML03P1 trial. We found no association between ITGA5 expression and clinical outcome. In contrast, patients with the highest relative expression (Q4) of the –E2/3 ITGA5 splice variant less likely had low‐risk disease than Q1–3 patients (21% vs. 38%, P = 0.027). Q4 patients had worse response to chemotherapy with a higher proportion having persistent minimal residual disease (50% vs. 23%, P = 0.003) and inferior overall survival (at 5 years: 48% vs. 67%, P = 0.015); the latter association was limited to low‐risk patients (Q4 vs. Q1‐3: 56% vs. 85%, P = 0.043) and was not seen in standard‐risk (51% vs. 60%, P = 0.340) or high‐risk (33% vs. 38%, P = 0.952) patients. Our exploratory studies indicate that transcriptome sequencing is useful for biomarker discovery, as exemplified by the identification of ITGA5 –E2/3 splice variant as potential novel adverse prognostic marker for low‐risk AML that, if confirmed, could serve to further risk‐stratify this patient subset. Am. J. Hematol. 88:694–702, 2013.


Leukemia | 2018

Simultaneous multiple interaction T-cell engaging (SMITE) bispecific antibodies overcome bispecific T-cell engager (BiTE) resistance via CD28 co-stimulation

Colin Correnti; George S. Laszlo; Willem J. de van der Schueren; Colin D. Godwin; Ashok D. Bandaranayake; Melanie A. Busch; Chelsea J. Gudgeon; Olivia M. Bates; James M. Olson; Christopher Mehlin; Roland B. Walter

The efficacy of early bispecific antibodies redirecting T-cells to eradicate cancer cells was partly limited because of suboptimal effector cell engagement.1 More efficient T-cell activation has been obtained with single-chain variable fragment (scFv) antibodies, notably Bispecific T-cell Engagers (BiTEs).2 Activity of the CD19/CD3 BiTE blinatumomab in adults and children with chemotherapy-resistant CD19+ B-cell acute lymphoblastic leukemia (B-ALL) led to regulatory drug approval in Europe and the United States. Many other BiTEs, all relying on CD3 signaling without providing co-stimulation, are in clinical testing in several solid tumors and hematologic malignancies.2,3


Leukemia & Lymphoma | 2013

High expression of neutrophil elastase predicts improved survival in pediatric acute myeloid leukemia: a report from the Children ' s Oncology Group

Chelsea J. Gudgeon; Kimberly H. Harrington; George S. Laszlo; Todd A. Alonzo; Robert B. Gerbing; Alan S. Gamis; Susana C. Raimondi; Betsy Hirsch; Soheil Meshinchi; Roland B. Walter

Neutrophil elastase, a serine protease of the chymotrypsin family encoded by ELANE (ELA2), is primarily found in azurophilic (primary) granules of neutrophils [1,2]. Physiologically, neutrophil elastase plays a role as an intracellular microbicidal agent that, after release, remains exposed at the cell surface and contributes to the extracellular killing of microorganisms; the protease also regulates immune responses by activating specific cell surface receptors and modulating the cytokine network [1,2].

Collaboration


Dive into the Chelsea J. Gudgeon's collaboration.

Top Co-Authors

Avatar

Roland B. Walter

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

George S. Laszlo

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Kimberly H. Harrington

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Soheil Meshinchi

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Alan S. Gamis

Children's Mercy Hospital

View shared research outputs
Top Co-Authors

Avatar

Betsy Hirsch

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Rhonda E. Ries

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susana C. Raimondi

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Todd A. Alonzo

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge