Network


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

Hotspot


Dive into the research topics where Esther de Boer is active.

Publication


Featured researches published by Esther de Boer.


Clinical Cancer Research | 2015

Safety and Tumor-specificity of Cetuximab-IRDye800 for Surgical Navigation in Head and Neck Cancer

Eben L. Rosenthal; Jason M. Warram; Esther de Boer; Thomas K. Chung; Melissa L. Korb; Margie Brandwein-Gensler; Theresa V. Strong; Cecelia E. Schmalbach; Anthony Morlandt; Garima Agarwal; Yolanda E. Hartman; William R. Carroll; Joshua S. Richman; Lisa Clemons; Lisle Nabell; Kurt R. Zinn

Purpose: Positive margins dominate clinical outcomes after surgical resections in most solid cancer types, including head and neck squamous cell carcinoma. Unfortunately, surgeons remove cancer in the same manner they have for a century with complete dependence on subjective tissue changes to identify cancer in the operating room. To effect change, we hypothesize that EGFR can be targeted for safe and specific real-time localization of cancer. Experimental Design: A dose escalation study of cetuximab conjugated to IRDye800 was performed in patients (n = 12) undergoing surgical resection of squamous cell carcinoma arising in the head and neck. Safety and pharmacokinetic data were obtained out to 30 days after infusion. Multi-instrument fluorescence imaging was performed in the operating room and in surgical pathology. Results: There were no grade 2 or higher adverse events attributable to cetuximab-IRDye800. Fluorescence imaging with an intraoperative, wide-field device successfully differentiated tumor from normal tissue during resection with an average tumor-to-background ratio of 5.2 in the highest dose range. Optical imaging identified opportunity for more precise identification of tumor during the surgical procedure and during the pathologic analysis of tissues ex vivo. Fluorescence levels positively correlated with EGFR levels. Conclusions: We demonstrate for the first time that commercially available antibodies can be fluorescently labeled and safely administered to humans to identify cancer with sub-millimeter resolution, which has the potential to improve outcomes in clinical oncology. Clin Cancer Res; 21(16); 3658–66. ©2015 AACR.


Scientific Reports | 2015

In Vivo Fluorescence Immunohistochemistry: Localization of Fluorescently Labeled Cetuximab in Squamous Cell Carcinomas

Esther de Boer; Jason M. Warram; Matthew D. Tucker; Yolanda E. Hartman; Lindsay S. Moore; Johannes S. de Jong; Thomas K. Chung; Melissa L. Korb; Kurt R. Zinn; Gooitzen M. van Dam; Eben L. Rosenthal; Margaret Brandwein-Gensler

Anti-EGFR (epidermal growth factor receptor) antibody based treatment strategies have been successfully implemented in head and neck squamous cell carcinoma (HNSCC). Unfortunately, predicting an accurate and reliable therapeutic response remains a challenge on a per-patient basis. Although significant efforts have been invested in understanding EGFR-mediated changes in cell signaling related to treatment efficacy, the delivery and histological localization in (peri-)tumoral compartments of antibody-based therapeutics in human tumors is poorly understood nor ever made visible. In this first in-human study of a systemically administered near-infrared (NIR) fluorescently labeled therapeutic antibody, cetuximab-IRDye800CW (2.5 mg/m2, 25 mg/m2, and 62.5 mg/m2), we show that by optical molecular imaging (i.e. denominated as In vivo Fluorescence Immunohistochemistry) we were able to evaluate localization of fluorescently labeled cetuximab. Clearly, optical molecular imaging with fluorescently labeled antibodies correlating morphological (peri-)tumoral characteristics to levels of antibody delivery, may improve treatment paradigms based on understanding true tumoral antibody delivery.


Cancer and Metastasis Reviews | 2014

Antibody-based imaging strategies for cancer

Jason M. Warram; Esther de Boer; Anna G. Sorace; Thomas K. Chung; Hyunki Kim; Rick G. Pleijhuis; Gooitzen M. van Dam; Eben L. Rosenthal

Although mainly developed for preclinical research and therapeutic use, antibodies have high antigen specificity, which can be used as a courier to selectively deliver a diagnostic probe or therapeutic agent to cancer. It is generally accepted that the optimal antigen for imaging will depend on both the expression in the tumor relative to normal tissue and the homogeneity of expression throughout the tumor mass and between patients. For the purpose of diagnostic imaging, novel antibodies can be developed to target antigens for disease detection, or current FDA-approved antibodies can be repurposed with the covalent addition of an imaging probe. Reuse of therapeutic antibodies for diagnostic purposes reduces translational costs since the safety profile of the antibody is well defined and the agent is already available under conditions suitable for human use. In this review, we will explore a wide range of antibodies and imaging modalities that are being translated to the clinic for cancer identification and surgical treatment.


Annals of Surgery | 2017

Oncologic Procedures Amenable to Fluorescence-guided Surgery

Kiranya E. Tipirneni; Jason M. Warram; Lindsay S. Moore; Andrew C. Prince; Esther de Boer; Aditi Jani; Irene Wapnir; Joseph C. Liao; Michael Bouvet; Nicole K. Behnke; Mary T. Hawn; George A. Poultsides; Alexander L. Vahrmeijer; William R. Carroll; Kurt R. Zinn; Eben L. Rosenthal

Objective: Although fluorescence imaging is being applied to a wide range of cancers, it remains unclear which disease populations will benefit greatest. Therefore, we review the potential of this technology to improve outcomes in surgical oncology with attention to the various surgical procedures while exploring trial endpoints that may be optimal for each tumor type. Background: For many tumors, primary treatment is surgical resection with negative margins, which corresponds to improved survival and a reduction in subsequent adjuvant therapies. Despite unfavorable effect on patient outcomes, margin positivity rate has not changed significantly over the years. Thus, patients often experience high rates of re-excision, radical resections, and overtreatment. However, fluorescence-guided surgery (FGS) has brought forth new light by allowing detection of subclinical disease not readily visible with the naked eye. Methods: We performed a systematic review of clinicatrials.gov using search terms “fluorescence,” “image-guided surgery,” and “near-infrared imaging” to identify trials utilizing FGS for those received on or before May 2016. Inclusion criteria: fluorescence surgery for tumor debulking, wide local excision, whole-organ resection, and peritoneal metastases. Exclusion criteria: fluorescence in situ hybridization, fluorescence imaging for lymph node mapping, nonmalignant lesions, nonsurgical purposes, or image guidance without fluorescence. Results: Initial search produced 844 entries, which was narrowed down to 68 trials. Review of literature and clinical trials identified 3 primary resection methods for utilizing FGS: (1) debulking, (2) wide local excision, and (3) whole organ excision. Conclusions: The use of FGS as a surgical guide enhancement has the potential to improve survival and quality of life outcomes for patients. And, as the number of clinical trials rise each year, it is apparent that FGS has great potential for a broad range of clinical applications.


Journal of Minimally Invasive Gynecology | 2015

Laparoscopic Fluorescent Visualization of the Ureter With Intravenous IRDye800CW

Melissa L. Korb; Warner K. Huh; Jonathan D. Boone; Jason M. Warram; Thomas K. Chung; Esther de Boer; Kirby I. Bland; Eben L. Rosenthal

OBJECTIVES Ureter injury is a serious complication of laparoscopic surgery. Current strategies to identify the ureters, such as placement of a ureteral stent, carry additional risks for patients. We hypothesize that the systemically injected near-infrared (NIR) dye IRDye800CW-CA can be used to visualize ureters intraoperatively. METHODS Adult female mixed-breed pigs weighing 24 to 41 kg (n = 2 per dose) were given a 30, 60, or 120 μg/kg systemic injection of IRDye800CW-CA. Using the Food and Drug Administration-cleared Pinpoint laparoscopic NIR system, images of the ureter and bladder were captured every 10 minutes for 60 minutes after injection. To determine the biodistribution of the dye, tissues were collected for ex vivo analysis with the Pearl Impulse system. ImageJ software was used to quantify fluorescence signal and signal-to-background ratio (SBR) for the intraoperative images. RESULTS The ureter was identified in all pigs at each dose, with peak intensity reached by 30 minutes and remaining elevated throughout the duration of imaging (60 minutes). The 60 μg/kg dose was determined to be optimal for differentiating ureters according to absolute fluorescence (>60 counts/pixel) and SBR (3.1). Urine fluorescence was inversely related to plasma fluorescence (R(2) = -0.82). Ex vivo imaging of kidney, ureter, bladder, and abdominal wall tissues revealed low fluorescence. CONCLUSION Systemic administration of IRDye800CW-CA shows promise in providing ureteral identification with high specificity during laparoscopic surgery. The low dose required, rapid time to visualization, and absence of invasive ureteral instrumentation inherent to this technique may reduce complications related to pelvic surgery.


British Journal of Neurosurgery | 2015

Fluorescence-guided resection of experimental malignant glioma using cetuximab-IRDye 800CW

Jason M. Warram; Esther de Boer; Melissa L. Korb; Yolanda E. Hartman; Joy L. Kovar; James M. Markert; G. Yancey Gillespie; Eben L. Rosenthal

The standard treatment for glioblastoma multiforme (GBM) remains maximal safe surgical resection. Here, we evaluated the ability of a systemically administered antibody–dye probe conjugate (cetuximab-IRDye 800CW) to provide sufficient fluorescent contrast for surgical resection of disease in both subcutaneous and orthotopic animal models of GBM. Multiple luciferase-positive GBM cell lines (D-54MG, U-87MG, and U-251MG; n = 5) were implanted in mouse flank and tumors were fluorescently imaged daily using a closed-field near-infrared (NIR) system after cetuximab-IRDye 800CW systemic administration. Orthotopic models were also generated (n = 5), and tumor resection was performed under white light and fluorescence guidance using an FDA-approved wide-field NIR imaging system. Residual tumor was monitored using luciferase imaging. Immunohistochemistry was performed to characterize tumor fluorescence, epidermal growth factor receptor (EGFR) expression, and vessel density. Daily imaging of tumors revealed an average tumor-to-background (TBR) of 4.5 for U-87MG, 4.1 for D-54MG, and 3.7 for U-251MG. Fluorescence intensity within the tumors peaked on day-1 after cetuximab-IRDye 800CW administration, however the TBR increased over time in two of the three cell lines. For the orthotopic model, TBR on surgery day ranged from 19 to 23 during wide-field, intraoperative imaging. Surgical resection under white light on day 3 after cetuximab-IRDye 800CW resulted in an average 41% reduction in luciferase signal while fluorescence-guided resection using wide-field NIR imaging resulted in a significantly (P = 0.001) greater reduction in luciferase signal (87%). Reduction of luciferase signal was found to correlate (R2 = 0.99) with reduction in fluorescence intensity. Fluorescence intensity was found to correlate (P < 0.05) with EGFR expression in D-54MG and U-251MG tumor types but not U-87MG. However, tumor fluorescence was found to correlate with vessel density for the U-87MG tumors. Here we show systemic administration of cetuximab-IRDye 800CW in combination with wide-field NIR imaging provided robust and specific fluorescence contrast for successful localization of disease in subcutaneous and orthotopic animal models of GBM.


Journal of Surgical Oncology | 2015

A ratiometric threshold for determining presence of cancer during fluorescence-guided surgery.

Jason M. Warram; Esther de Boer; Lindsay S. Moore; Cecelia E. Schmalbach; Kirk P. Withrow; William R. Carroll; Joshua S. Richman; Anthony Morlandt; Margaret Brandwein-Gensler; Eben L. Rosenthal

Fluorescence‐guided imaging to assist in identification of malignant margins has the potential to dramatically improve oncologic surgery. However, a standardized method for quantitative assessment of disease‐specific fluorescence has not been investigated. Introduced here is a ratiometric threshold derived from mean fluorescent tissue intensity that can be used to semi‐quantitatively delineate tumor from normal tissue.


The Journal of Pathology: Clinical Research | 2016

Fluorescence imaging to localize head and neck squamous cell carcinoma for enhanced pathological assessment

Jason M. Warram; Esther de Boer; Gooitzen M. van Dam; Lindsay S. Moore; Stephanie L. Bevans; Erika M. Walsh; Erik Scott Young; William R. Carroll; Todd M. Stevens; Eben L. Rosenthal

Accurately identifying close or positive margins in real‐time permits re‐excision during surgical procedures. Intraoperative assessment of margins via gross examination and frozen section is a widely used tool to assist the surgeon in achieving complete resection. While this methodology permits diagnosis of freshly resected tissue, the process is fraught with misinterpretation and sampling errors. During fluorescence‐guided surgery, an exogenous fluorescent agent specific for the target disease is imaged in order to navigate the surgical excision. As this technique quickly advances into the clinic, we hypothesize that the disease‐specific fluorescence inherently contained within the resected tissues can be used to guide histopathological assessment. To evaluate the feasibility of fluorescence‐guided pathology, we evaluated head and neck squamous cell carcinoma tumour specimens and margins resected from animals and patients after systemic injection of cetuximab‐IRDye800CW. In a preclinical model of luciferase‐positive tumour resection using bioluminescence as the gold standard, fluorescence assessment determined by closed‐field fluorescence imaging of fresh resected margins accurately predicted the presence of disease in 33/39 positive margins yielding an overall sensitivity of 85%, specificity of 95%, positive predictive value (PPV) of 94%, and a negative predictive value (NPV) of 87%, which was superior to both surgical assessment (54%, 61%, 57%, and 58%) and pathological assessment (49%, 95%, 91%, and 66%), respectively. When the power of the technique was evaluated using human‐derived tumour tissues, as little as 0.5mg (1mm3) of tumour tissue was identified (tumour‐to‐background‐ratio:5.2). When the sensitivity/specificity of fluorescence‐guided pathology was determined using traditional histological assessment as the gold standard in human tissues obtained during fluorescence‐guided surgery, the technique was highly accurate with a sensitivity of 91%, specificity of 85%, PPV of 81%, and NPV of 93% for 90 human‐derived samples. This approach can be used as a companion to the pathologist, eliminating confounding factors while impacting surgical intervention and patient management.


The Journal of Nuclear Medicine | 2017

Characterizing the Utility and Limitations of Repurposing an Open-Field Optical Imaging Device for Fluorescence-Guided Surgery in Head and Neck Cancer Patients

Lindsay S. Moore; Eben L. Rosenthal; Thomas K. Chung; Esther de Boer; Neel Patel; Andrew C. Prince; Melissa L. Korb; Erika M. Walsh; E. Scott Young; Todd M. Stevens; Kirk P. Withrow; Anthony Morlandt; Joshua S. Richman; William R. Carroll; Kurt R. Zinn; Jason M. Warram

The purpose of this study was to assess the potential of U.S. Food and Drug Administration–cleared devices designed for indocyanine green–based perfusion imaging to identify cancer-specific bioconjugates with overlapping excitation and emission wavelengths. Recent clinical trials have demonstrated potential for fluorescence-guided surgery, but the time and cost of the approval process may impede clinical translation. To expedite this translation, we explored the feasibility of repurposing existing optical imaging devices for fluorescence-guided surgery. Methods: Consenting patients (n = 15) scheduled for curative resection were enrolled in a clinical trial evaluating the safety and specificity of cetuximab-IRDye800 (NCT01987375). Open-field fluorescence imaging was performed preoperatively and during the surgical resection. Fluorescence intensity was quantified using integrated instrument software, and the tumor-to-background ratio characterized fluorescence contrast. Results: In the preoperative clinic, the open-field device demonstrated potential to guide preoperative mapping of tumor borders, optimize the day of surgery, and identify occult lesions. Intraoperatively, the device demonstrated robust potential to guide surgical resections, as all peak tumor-to-background ratios were greater than 2 (range, 2.2–14.1). Postresection wound bed fluorescence was significantly less than preresection tumor fluorescence (P < 0.001). The repurposed device also successfully identified positive margins. Conclusion: The open-field imaging device was successfully repurposed to distinguish cancer from normal tissue in the preoperative clinic and throughout surgical resection. This study illuminated the potential for existing open-field optical imaging devices with overlapping excitation and emission spectra to be used for fluorescence-guided surgery.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2016

On the horizon: Optical imaging for cutaneous squamous cell carcinoma

Esther de Boer; Lindsay S. Moore; Jason M. Warram; Conway C. Huang; Margaret Brandwein-Gensler; Gooitzen M. van Dam; Eben L. Rosenthal; Cecelia E. Schmalbach

Surgical resection with negative margins remains the standard of care for high‐risk cutaneous squamous cell carcinoma (SCC). However, surgical management is often limited by poor intraoperative tumor visualization and inability to detect occult nodal metastasis. The inability to intraoperatively detect microscopic disease can lead to additional surgery, tumor recurrence, and decreased survival.

Collaboration


Dive into the Esther de Boer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason M. Warram

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Lindsay S. Moore

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Melissa L. Korb

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

William R. Carroll

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gooitzen M. van Dam

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Kurt R. Zinn

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Thomas K. Chung

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Anthony Morlandt

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Margaret Brandwein-Gensler

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge