Lars Wagner

Overview:

I am a pediatric oncologist with a clinical and research focus on the care of adolescents and young adults with sarcoma. I have led multiple institutional and national clinical trials exploring new therapies for patients with recurrent sarcoma, and am interested in new drug development and identification of predictive biomarkers.

Positions:

Professor of Pediatrics

Pediatrics, Hematology-Oncology
School of Medicine

Chief, Division of Hematology/Oncology

Pediatrics, Hematology-Oncology
School of Medicine

Member of the Duke Cancer Institute of the

Duke Cancer Institute
School of Medicine

Education:

M.D. 1991

University of Kentucky, College of Medicine

Residency, Pediatrics

University of Tennessee Health Science Center

Hematology/Oncology Fellowship, St Jude Children's Hospital

University of Tennessee Health Science Center

Grants:

Per Case Reimbursement : NIH National Clinical Trials Network (NCTN)

Administered By
Pediatrics, Hematology-Oncology
Awarded By
Children's Hospital of Philadelphia
Role
Principal Investigator
Start Date
End Date

A Multi-Center Phase III, Randomized, Open-Label Trial of Vigil (bi-shRNAfurin and GMCSF Augmented Autologous Tumor Cell Immunotherapy) in combination with Irinotecan and Temozolomide as a Second-Line Regimen for Ewing's Sarcoma (CL-PTL-130)

Administered By
Pediatrics, Hematology-Oncology
Awarded By
Gradalis, Inc.
Role
Principal Investigator
Start Date
End Date

Workload Intensity NIH National Clinical Trials Network (NCTN)

Administered By
Pediatrics, Hematology-Oncology
Awarded By
Children's Hospital of Philadelphia
Role
Principal Investigator
Start Date
End Date

NCI Community Oncology Research Program (NCORP) Research Base Grant

Administered By
Pediatrics, Hematology-Oncology
Awarded By
Children's Hospital of Philadelphia
Role
Principal Investigator
Start Date
End Date

Project: One Time EOY, Integrated BIQSFP AAML1031- Echocardiogram Reports and Images

Administered By
Pediatrics, Hematology-Oncology
Awarded By
Children's Hospital of Philadelphia
Role
Principal Investigator
Start Date
End Date

Publications:

Sarcoma as the Prototype Disease for Adolescent and Young Adult Programs.

Authors
MLA Citation
Wagner, Lars M. “Sarcoma as the Prototype Disease for Adolescent and Young Adult Programs.J Oncol Pract, vol. 15, no. 5, May 2019, pp. 249–50. Pubmed, doi:10.1200/JOP.19.00145.
URI
https://scholars.duke.edu/individual/pub1386036
PMID
31075213
Source
pubmed
Published In
J Oncol Pract
Volume
15
Published Date
Start Page
249
End Page
250
DOI
10.1200/JOP.19.00145

Phase I trial of weekly MK-0752 in children with refractory central nervous system malignancies: a pediatric brain tumor consortium study.

PURPOSE: Amplification and high levels of NOTCH ligand expression have been identified in several types of pediatric brain tumors. A phase I trial of weekly MK-0752, an oral inhibitor of gamma-secretase, was conducted in children with recurrent central nervous system (CNS) malignancies to estimate the maximum tolerated dose, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics of weekly MK-0752. METHODS: MK-0752 was administered once weekly at 1000 and 1400 mg/m(2) using a rolling-6 design. PK analysis was performed during the first course. NOTCH and HES expression was assessed by immunohistochemistry and Western blot. RESULTS: Ten eligible patients were enrolled (median age 8.8 years; range 3.1-19.2) with diagnoses of brain stem glioma (n = 3), ependymoma (n = 2), anaplastic astrocytoma (n = 1), choroid plexus carcinoma (n = 2), medulloblastoma (n = 1), and primitive neuroectodermal tumor (n = 1). Nine were evaluable for toxicity. One DLT of fatigue occurred in the six evaluable patients enrolled at 1000 mg/m(2)/dose. No DLTs were experienced by three patients treated at 1400 mg/m(2)/dose. Non-dose-limiting grade 3 toxicities included lymphopenia, neutropenia, and anemia. Median number of treatment courses was 2 (range 1-10). Two patients continued on therapy for at least 6 months. The median (range) C(max) of MK-0752 was 88.2 μg/mL (40.6 to 109 μg/mL) and 60.3 μg/mL (59.2 to 91.9 μg/mL) in patients receiving 1000 and 1400 mg/m(2)/week, respectively. NOTCH expression was decreased in six of seven patients for whom tissue was available at 24 h post-MK-0752. CONCLUSION: MK-0752 is well tolerated and exhibits target inhibition at 1000 and 1400 mg/m(2)/week in children with recurrent CNS malignancies.
Authors
Hoffman, LM; Fouladi, M; Olson, J; Daryani, VM; Stewart, CF; Wetmore, C; Kocak, M; Onar-Thomas, A; Wagner, L; Gururangan, S; Packer, RJ; Blaney, SM; Gajjar, A; Kun, LE; Boyett, JM; Gilbertson, RJ
MLA Citation
Hoffman, Lindsey M., et al. “Phase I trial of weekly MK-0752 in children with refractory central nervous system malignancies: a pediatric brain tumor consortium study.Childs Nerv Syst, vol. 31, no. 8, Aug. 2015, pp. 1283–89. Pubmed, doi:10.1007/s00381-015-2725-3.
URI
https://scholars.duke.edu/individual/pub1085440
PMID
25930724
Source
pubmed
Published In
Childs Nerv Syst
Volume
31
Published Date
Start Page
1283
End Page
1289
DOI
10.1007/s00381-015-2725-3

Thyroid Nodules on Ultrasound in Children and Young Adults: Comparison of Diagnostic Performance of Radiologists' Impressions, ACR TI-RADS, and a Deep Learning Algorithm.

BACKGROUND. In current clinical practice, thyroid nodules in children are generally evaluated on the basis of radiologists' overall impressions of ultrasound images. OBJECTIVE. The purpose of this article is to compare the diagnostic performance of radiologists' overall impression, the American College of Radiology Thyroid Imaging Reporting and Data System (ACR TI-RADS), and a deep learning algorithm in differentiating benign and malignant thyroid nodules on ultrasound in children and young adults. METHODS. This retrospective study included 139 patients (median age 17.5 years; 119 female patients, 20 male patients) evaluated from January 1, 2004, to September 18, 2020, who were 21 years old and younger with a thyroid nodule on ultrasound with definitive pathologic results from fine-needle aspiration and/or surgical excision to serve as the reference standard. A single nodule per patient was selected, and one transverse and one longitudinal image each of the nodules were extracted for further evaluation. Three radiologists independently characterized nodules on the basis of their overall impression (benign vs malignant) and ACR TI-RADS. A previously developed deep learning algorithm determined for each nodule a likelihood of malignancy, which was used to derive a risk level. Sensitivities and specificities for malignancy were calculated. Agreement was assessed using Cohen kappa coefficients. RESULTS. For radiologists' overall impression, sensitivity ranged from 32.1% to 75.0% (mean, 58.3%; 95% CI, 49.2-67.3%), and specificity ranged from 63.8% to 93.9% (mean, 79.9%; 95% CI, 73.8-85.7%). For ACR TI-RADS, sensitivity ranged from 82.1% to 87.5% (mean, 85.1%; 95% CI, 77.3-92.1%), and specificity ranged from 47.0% to 54.2% (mean, 50.6%; 95% CI, 41.4-59.8%). The deep learning algorithm had a sensitivity of 87.5% (95% CI, 78.3-95.5%) and specificity of 36.1% (95% CI, 25.6-46.8%). Interobserver agreement among pairwise combinations of readers, expressed as kappa, for overall impression was 0.227-0.472 and for ACR TI-RADS was 0.597-0.643. CONCLUSION. Both ACR TI-RADS and the deep learning algorithm had higher sensitivity albeit lower specificity compared with overall impressions. The deep learning algorithm had similar sensitivity but lower specificity than ACR TI-RADS. Interobserver agreement was higher for ACR TI-RADS than for overall impressions. CLINICAL IMPACT. ACR TI-RADS and the deep learning algorithm may serve as potential alternative strategies for guiding decisions to perform fine-needle aspiration of thyroid nodules in children.
Authors
Yang, J; Page, LC; Wagner, L; Wildman-Tobriner, B; Bisset, L; Frush, D; Mazurowski, MA
MLA Citation
Yang, Jichen, et al. “Thyroid Nodules on Ultrasound in Children and Young Adults: Comparison of Diagnostic Performance of Radiologists' Impressions, ACR TI-RADS, and a Deep Learning Algorithm.Ajr Am J Roentgenol, vol. 220, no. 3, Mar. 2023, pp. 408–17. Pubmed, doi:10.2214/AJR.22.28231.
URI
https://scholars.duke.edu/individual/pub1554606
PMID
36259591
Source
pubmed
Published In
Ajr. American Journal of Roentgenology
Volume
220
Published Date
Start Page
408
End Page
417
DOI
10.2214/AJR.22.28231

In situ mass spectrometry imaging reveals heterogeneous glycogen stores in human normal and cancerous tissues.

Glycogen dysregulation is a hallmark of aging, and aberrant glycogen drives metabolic reprogramming and pathogenesis in multiple diseases. However, glycogen heterogeneity in healthy and diseased tissues remains largely unknown. Herein, we describe a method to define spatial glycogen architecture in mouse and human tissues using matrix-assisted laser desorption/ionization mass spectrometry imaging. This assay provides robust and sensitive spatial glycogen quantification and architecture characterization in the brain, liver, kidney, testis, lung, bladder, and even the bone. Armed with this tool, we interrogated glycogen spatial distribution and architecture in different types of human cancers. We demonstrate that glycogen stores and architecture are heterogeneous among diseases. Additionally, we observe unique hyperphosphorylated glycogen accumulation in Ewing sarcoma, a pediatric bone cancer. Using preclinical models, we correct glycogen hyperphosphorylation in Ewing sarcoma through genetic and pharmacological interventions that ablate in vivo tumor growth, demonstrating the clinical therapeutic potential of targeting glycogen in Ewing sarcoma.
Authors
Young, LEA; Conroy, LR; Clarke, HA; Hawkinson, TR; Bolton, KE; Sanders, WC; Chang, JE; Webb, MB; Alilain, WJ; Vander Kooi, CW; Drake, RR; Andres, DA; Badgett, TC; Wagner, LM; Allison, DB; Sun, RC; Gentry, MS
MLA Citation
Young, Lyndsay E. A., et al. “In situ mass spectrometry imaging reveals heterogeneous glycogen stores in human normal and cancerous tissues.Embo Mol Med, vol. 14, no. 11, Nov. 2022, p. e16029. Pubmed, doi:10.15252/emmm.202216029.
URI
https://scholars.duke.edu/individual/pub1546604
PMID
36059248
Source
pubmed
Published In
Embo Mol Med
Volume
14
Published Date
Start Page
e16029
DOI
10.15252/emmm.202216029

Immunotherapy for osteosarcoma: Where do we go from here?

Osteosarcoma is the most common bone tumor in children and young adults, with few advances in survival and treatment, especially for metastatic disease, in the last 30 years. Recently, immunotherapy has begun to show promise in various adult cancers, but the utility of this approach for osteosarcoma remains relatively unexplored. In this review, we outline the mechanisms and status of immunotherapies currently in clinical trials as well as future therapies on the horizon, and discuss their potential application for osteosarcoma.
Authors
Wedekind, MF; Wagner, LM; Cripe, TP
MLA Citation
Wedekind, Mary F., et al. “Immunotherapy for osteosarcoma: Where do we go from here?Pediatr Blood Cancer, vol. 65, no. 9, Sept. 2018, p. e27227. Pubmed, doi:10.1002/pbc.27227.
URI
https://scholars.duke.edu/individual/pub1530365
PMID
29923370
Source
pubmed
Published In
Pediatr Blood Cancer
Volume
65
Published Date
Start Page
e27227
DOI
10.1002/pbc.27227