Thick tissue diffusion model with binding to optimize topical staining in fluorescence breast cancer margin imaging

Intraoperative tumor/surgical margin assessment is required to achieve higher tumor resection rate in breast-conserving surgery. Though current histology provides incomparable accuracy in margin assessment, thin tissue sectioning and the limited field of view of microscopy makes histology too time-consuming for intraoperative applications. If thick tissue, wide-field imaging can provide an acceptable assessment of tumor cells at the surface of resected tissues, an intraoperative protocol can be developed to guide the surgery and provide immediate feedback for surgeons. Topical staining of margins with cancer-targeted molecular imaging agents has the potential to provide the sensitivity needed to see microscopic cancer on a wide-field image; however, diffusion and nonspecific retention of imaging agents in thick tissue can significantly diminish tumor contrast with conventional methods. Here, we present a mathematical model to accurately simulate nonspecific retention, binding, and diffusion of imaging agents in thick tissue topical staining to guide and optimize future thick tissue staining and imaging protocol. In order to verify the accuracy and applicability of the model, diffusion profiles of cancer targeted and untargeted (control) nanoparticles at different staining times in A431 tumor xenografts were acquired for model comparison and tuning. The initial findings suggest the existence of nonspecific retention in the tissue, especially at the tissue surface. The simulator can be used to compare the effect of nonspecific retention, receptor binding and diffusion under various conditions (tissue type, imaging agent) and provides optimal staining and imaging protocols for targeted and control imaging agent.

[1]  Xiaochun Xu,et al.  Rinsing paired-agent model (RPAM) to quantify cell-surface receptor concentrations in topical staining applications of thick tissues , 2017, Physics in medicine and biology.

[2]  K. Geisinger,et al.  Intraoperative evaluation of lumpectomy margins by imprint cytology with histologic correlation: a community hospital experience. , 2002, Archives of pathology & laboratory medicine.

[3]  Ting-Yim Lee,et al.  An Adiabatic Approximation to the Tissue Homogeneity Model for Water Exchange in the Brain: I. Theoretical Derivation , 1998, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[4]  Adam K. Glaser,et al.  Multiplexed Optical Imaging of Tumor-Directed Nanoparticles: A Review of Imaging Systems and Approaches , 2017, Nanotheranostics.

[5]  L. Jacobs,et al.  Positive Margins: The Challenge Continues for Breast Surgeons , 2008, Annals of Surgical Oncology.

[6]  A. Lammertsma,et al.  Simplified Reference Tissue Model for PET Receptor Studies , 1996, NeuroImage.

[7]  Jovan G. Brankov,et al.  Quantification of the binding potential of cell-surface receptors in fresh excised specimens via dual-probe modeling of SERS nanoparticles , 2015, Scientific Reports.

[8]  A. Jemal,et al.  Cancer statistics, 2014 , 2014, CA: a cancer journal for clinicians.

[9]  Venkataramanan Krishnaswamy,et al.  Spectral discrimination of breast pathologies in situ using spatial frequency domain imaging , 2013, Breast Cancer Research.

[10]  Christilyn P. Graff,et al.  Theoretical analysis of antibody targeting of tumor spheroids: importance of dosage for penetration, and affinity for retention. , 2003, Cancer research.

[11]  Brian C Wilson,et al.  Rapid ratiometric biomarker detection with topically applied SERS nanoparticles. , 2014, Technology.

[12]  D. Wruk,et al.  Accuracy of Frozen Section Analysis Versus Specimen Radiography During Breast-Conserving Surgery for Nonpalpable Lesions , 2008, World Journal of Surgery.

[13]  L. Newman,et al.  Predictors of Re-excision among Women Undergoing Breast-Conserving Surgery for Cancer , 2008, Annals of Surgical Oncology.

[14]  Freddy T. Nguyen,et al.  Intraoperative evaluation of breast tumor margins with optical coherence tomography. , 2009, Cancer research.

[15]  Xiaochun Xu,et al.  Microscopic investigation of" topically applied nanoparticles for molecular imaging of fresh tissue surfaces , 2018, Journal of biophotonics.

[16]  M. Trivella,et al.  Reoperation rates after breast conserving surgery for breast cancer among women in England: retrospective study of hospital episode statistics , 2012, BMJ : British Medical Journal.

[17]  M. Nakano,et al.  Efficacy of intraoperative entire-circumferential frozen section analysis of lumpectomy margins during breast-conserving surgery for breast cancer , 2015, International Journal of Clinical Oncology.

[18]  M Moschetta,et al.  Role of specimen US for predicting resection margin status in breast conserving therapy. , 2015, Il Giornale di chirurgia.

[19]  Adam K Glaser,et al.  Raman-Encoded Molecular Imaging with Topically Applied SERS Nanoparticles for Intraoperative Guidance of Lumpectomy. , 2017, Cancer research.

[20]  Milind Rajadhyaksha,et al.  Quantitative molecular phenotyping with topically applied SERS nanoparticles for intraoperative guidance of breast cancer lumpectomy , 2016, Scientific Reports.

[21]  Scott C. Davis,et al.  Optimizing fresh specimen staining for rapid identification of tumor biomarkers during surgery , 2017, Theranostics.

[22]  Scott C Davis,et al.  Topical dual-stain difference imaging for rapid intra-operative tumor identification in fresh specimens. , 2013, Optics letters.

[23]  M. Ramos,et al.  Ultrasound-guided excision combined with intraoperative assessment of gross macroscopic margins decreases the rate of reoperations for non-palpable invasive breast cancer. , 2013, Breast.

[24]  T. Smith,et al.  The efficacy of specimen radiography in evaluating the surgical margins of impalpable breast carcinoma. , 1994, AJR. American journal of roentgenology.