Bio


Quan is an Instructor in the Department of Neurosurgery and the Molecular Imaging Program at Stanford (MIPS), where she leads a Phase I/II clinical trial on molecular targeted fluorescence-guided surgery to improve resection accuracy of brain malignancies. She has established a clinical theranostic imaging and analysis framework to predict therapeutic distribution in brain tumors from clinical, radiographic and tumor microenvironment features. Her resesarch interests also include molecular imaging of intratumoral immunosuppression of high-grade glioma and enhanced antibody delivery across the blood-brain barrier.

Dr. Zhou studied Applied Biology and Chemistry on a four-year full ride scholarship at Hong Kong Baptist University. She obtained her MS and PhD in Biomedical Engineering at the University of Michigan, Ann Arbor, where she was awarded the Richard and Eleanor Towner Prize for Distinguished Academic Achievement. Dr. Zhou’s doctoral research on optical and photoacoustic imaging for early detection of liver and colorectal cancers with molecular targeting probes received the American Gastroenterological Association (AGA) Research Foundation Award.

Dr. Zhou has served as a mentor and counselor for students of all ages and backgrounds. In 2017 she became the first non-faculty recipient of Willie Hobbs Moore Aspire Advance Achieve Mentoring Award recognizing her outstanding mentorship in multiple academic, outreach and athletic programs, and her leadership roles in the Society of Women Engineers (SWE), the engineering honor society Tau Beta Pi (TBP) and the University of Michigan Table Tennis Club (UMTTC). At Stanford, she has been a key organizer for the MIPS Retreat and Journal Club.

Academic Appointments


Honors & Awards


  • Pilot Grant (PI), Maternal & Child Health Research Institute (MCHRI) (2024-2025)
  • Rising Star Award, Women in Molecular Imaging Network (WIMIN), World Molecular Imaging Society (2023)
  • Editorial Scholar, Society for Neuro-Oncology (SNO) and Oxford University Press (2023-2024)
  • Alavi-Mandell Award, Society of Nuclear Medicine and Molecular Imaging (SNMMI) (2023)
  • Scholar Award, Women in Molecular Imaging Network (WIMIN), World Molecular Imaging Society (2022, 2023)
  • National Cancer Institute (NCI) Grant, World Molecular Imaging Congress (WMIC) (2022)
  • Cover Article, July issue of Theranostics (2021)
  • Best Scientific Image Award, Molecular Imaging Program at Stanford (MIPS) (2018)
  • 1st Prize Poster Award, Molecular Imaging Program at Stanford (MIPS) (2018)
  • Research Foundation Award, American Gastroenterological Association (AGA) (2017)
  • Richard and Eleanor Towner Prize for Distinguished Academic Achievement, University of Michigan, Ann Arbor (2017)
  • Willie Hobbs Moore Aspire, Advance, Achieve Mentoring Award, University of Michigan, Ann Arbor (2017)
  • Rackham Graduate Student Research Grant, University of Michigan, Ann Arbor (2017)
  • Poster of Distinction, Digestive Disease Week (DDW), Chicago, IL (2017)
  • State Champion, National Collegiate Table Tennis Association (NCTTA), MI Division (2015-2017)
  • Most Outstanding Active Member, Tau Beta Pi Engineering Honor Society, Michigan Gamma Chapter (2013-2014)
  • Rackham International Travel Grant, University of Michigan, Ann Arbor (2011)
  • Summer Undergraduate Research Fellowship (SURF), Hong Kong Baptist University (2010)
  • Professor Herbert Tsang Scholarships, Hong Kong Baptist University (2008–2010)
  • Sir Tseng Chi Lu Scholarship, Hong Kong Baptist University (2007–2008)
  • Merit Prize, Short Story Writing Competition, Hong Kong Baptist University (2007)
  • Full Ride Scholarship for Outstanding Mainland Students, Education Bureau, Hong Kong (2006-2010)

Boards, Advisory Committees, Professional Organizations


  • Co-Chair, Women in Molecular Imaging Network (WIMIN), World Molecular Imaging Society (2024 - Present)
  • Fundraising/Auction Committee, Women in Molecular Imaging Network (WIMIN), World Molecular Imaging Society (2023 - Present)
  • Member, DEI Workgroup, World Molecular Imaging Society (WMIS) (2023 - Present)
  • Co-Lead, MIPS Trainees Council (MTC), Stanford (2022 - Present)
  • Reviewer, Session Chair & Moderator, Instrumentation Category, World Molecular Imaging Congress (WMIC) (2022 - 2022)
  • Guest Editor, Special Issue “Optical and Acoustical Methods for Biomedical Imaging and Sensing”, Sensors (2022 - Present)
  • Reviewer, Biomedical Engineering Society (BMES) Annual Meeting (2022 - 2022)
  • Alumni Mentor for Career Development, College of Engineering, University of Michigan, Ann Arbor (2021 - 2022)
  • Program Committee, Molecular Imaging Program at Stanford (MIPS) Retreat (2020 - 2020)
  • Organizer, Molecular Imaging Program at Stanford (MIPS) Journal Club (2018 - Present)
  • Secretary, Tau Beta Pi National Engineering Honor Society, MI-Gamma Chapter (2013 - 2014)
  • Co-Founder, Treasurer & Safety Officer, University of Michigan Table Tennis Club (2014 - 2017)
  • Treasurer, Graduate Society of Women Engineers, University of Michigan, Ann Arbor (2014 - 2015)
  • International Student Ambassador, Rackham Graduate School, University of Michigan, Ann Arbor (2012 - 2015)
  • Professional Development Chair, Graduate Society of Women Engineers, University of Michigan, Ann Arbor (2012 - 2014)
  • Member, World Molecular Imaging Society (WMIS) (2018 - Present)
  • Member, American Gastroenterological Association (2017 - Present)
  • Member, SPIE international society for optics and photonic (2017 - Present)
  • Member, The Optical Society (2016 - Present)
  • Member, Phi Kappa Phi National All-Discipline Honor Society (2014 - Present)

Professional Education


  • Doctor of Philosophy, University of Michigan, Ann Arbor, Biomedical Engineering (Biomedical Imaging track) (2017)
  • Master of Science, University of Michigan, Ann Arbor, Biomedical Engineering (Biomaterials track) (2012)
  • Bachelor of Science (Hons.), Hong Kong Baptist University, Applied Biology (Biotechnology) and Chemistry (2010)

Current Research and Scholarly Interests


Current Research Focus: molecular targeted theranostic imaging of brain tumor and enhanced drug delivery

Areas of Insterests: molecular imaging, theranostics, fluorescence-guided surgery, brain tumor, drug delivery

Dr. Zhou has made substantial contributions to the growing biomedical research field of Molecular Imaging. Molecular imaging emerged in the mid twentieth century as a highly specialized discipline at the intersection of molecular biology and in vivo imaging, focusing on imaging molecules of medical interest within intact living subjects. Dr. Zhou’s research addresses some of the nation’s most pressing issues related to the development of effective approaches for accurate detection of human diseases and improving their treatment outcome. Her innovations in molecular imaging technology enables the visualization, characterization, and quantification of biologic processes taking place at the cellular and subcellular levels. The multiple and numerous potentialities of Quan’s work are applicable to the diagnosis of diseases such as cancer, neurological and cardiovascular diseases. Her strong education background in biological sciences and biomedical engineering followed by postdoctoral training in translational and clinical research have helped her develop multiple disease-specific molecular probes and imaging strategies for early cancer diagnosis, image-guided surgery, therapeutic delivery prediction and at-risk cardiovascular plaque detection. Her research also contributes to improving the treatment of these disorders by testing and optimizing the execution of new interventions. Her work is expected to have a major economic impact due to earlier disease detection and personalized therapy.

Dr. Zhou’s research has led to emergence of novel solutions and opportunities, in particular, for molecular imaging of cancer and other diseases, for discovering, leveraging and integration of cancer biomarker and tumor microenvironment information, and for novel approaches to acquire real-time high-resolution contrast enhanced visualization of tumor margin and optimization based on imaging depth, quality and speed. Dr. Zhou has been able to formulate the involved clinical and biological problems into biomedical engineering frameworks and find ways to exploit a variety of modern techniques and approaches from photoacoustic imaging, fluorescence-guided surgery, micro-electromechanical systems and therapeutic delivery strategies in developing elegant and effective solutions. Her work in the Neurosurgery Department and Molecular Imaging Program at Stanford involves research related to developing tumor-specific molecular probes, advanced imaging methods and therapeutic delivery systems for adult and pediatric patients with malignant brain cancers to improve margin detection, enhance resection accuracy, and improve treatment outcome.

Clinical Trials


  • Panitumumab-IRDye800 in Diagnosing Participants With Malignant Glioma Undergoing Surgery Recruiting

    The phase I/II trial studies the side effects and best dose of panitumumab-IRDye800 in diagnosing participants with malignant glioma who undergo surgery. Panitumumab-IRDye800 can attach to tumor cells and make them more visible using a special camera during surgery, which may help surgeons better distinguish tumor cells from normal brain tissue and identify small tumors that cannot be seen using current imaging methods.

    View full details

  • Panitumumab-IRDye800 to Detect Pediatric Neoplasms During Neurosurgical Procedures Not Recruiting

    The objective of the study is to assess safety of panitumumab-IRDye800 in pediatric patients undergoing brain surgery to remove suspected tumors.

    Stanford is currently not accepting patients for this trial.

    View full details

All Publications


  • Factors for differential outcome across cancers in clinical molecular-targeted fluorescence imaging. Journal of nuclear medicine : official publication, Society of Nuclear Medicine Zhou, Q., van den Berg, N. S., Kang, W., Pei, J., Nishio, N., van Keulen, S., Engelen, M. A., Lee, Y. J., Hom, M., Vega Leonel, J. C., Hart, Z., Vogel, H., Cayrol, R., Martin, B. A., Roesner, M., Shields, G., Lui, N., Hayden Gephart, M., Raymundo, R. C., Yi, G., Granucci, M., Grant, G. A., Li, G., Rosenthal, E. L. 2022

    Abstract

    Clinical imaging performance using a fluorescent antibody was compared across three cancers to elucidate physical and biological factors contributing to differential translation of epidermal growth factor receptor (EGFR) expression to macroscopic fluorescence in tumors. Methods: Thirty-one patients with high-grade glioma (HGG, n = 5), head-and-neck squamous cell carcinoma (HNSCC, n = 23) or lung adenocarcinoma (LAC, n = 3) were systemically infused with 50 mg panitumumab-IRDye800, 1 - 3 days prior to surgery. Intraoperative open-field fluorescent images of the surgical field were acquired, where imaging device settings and operating room lighting conditions were tested on tissue-mimicking phantoms. Fluorescence contrast and margin size were measured on resected specimen surface. Antibody distribution and EGFR immunoreactivity were characterized in macroscopic and microscopic histological structures. Integrity of the blood-brain barrier (BBB) was examined via tight junction protein (claudin-5) expression with immunohistochemistry. Stepwise multivariate linear regression of biological variables was performed to identify independent predictors of panitumumab-IRDye800 concentration in tissue. Results: Optimally acquired at the lowest gain for tumor detection with ambient light, intraoperative fluorescence imaging enhanced tissue-size dependent tumor contrast by 5.2-fold, 3.4-fold and 1.4-fold in HGG, HNSCC and LAC, respectively. Tissue surface fluorescence target-to-background ratio correlated with margin size and identified 78 - 97% of at-risk resection margins ex vivo. In 4 µm-thick tissue sections, fluorescence detected tumor with 0.85 - 0.89 areas under the receiver operating characteristic curves. Preferential breakdown of BBB in HGG improved tumor specificity of intratumoral antibody distribution relative to that of EGFR (96% vs 80%) despite its reduced concentration (3.9 ng/mg tissue) compared to HNSCC (8.1 ng/mg) and LAC (6.3 ng/mg). Cellular EGFR expression, tumor cell density, plasma antibody concentration and delivery barrier were independently associated with local intratumoral panitumumab-IRDye800 concentration with 0.62 goodness-of-fit of prediction. Conclusion: In multi-cancer clinical imaging of receptor-ligand based molecular probe, plasma antibody concentration, delivery barrier, as well as intratumoral EGFR expression driven by cellular biomarker expression and tumor cell density, led to heterogeneous intratumoral antibody accumulation and spatial distribution while tumor size, resection margin, and intraoperative imaging settings substantially influenced macroscopic tumor contrast.

    View details for DOI 10.2967/jnumed.121.263674

    View details for PubMedID 35332092

  • Fluorescence-guided craniotomy of glioblastoma using panitumumab-IRDye800. Neurosurgical focus: Video Zhou, Q., Li, G. 2022; 6 (1): V9

    Abstract

    A contrast-enhancing lesion in the left temporal lobe of a 72-year-old woman was biopsied and diagnosed as glioblastoma. Near-infrared (NIR)-labeled epidermal growth factor receptor (EGFR) antibody, panitumumab-IRDye800, was infused 52 hours before craniotomy without pretreatment. Tumor fluorescence was detected through intact dura, and the visual contrast between disease and peritumoral healthy brain was enhanced after tumor exposure. Residual cancerous tissue was identified with strong fluorescence in resection cavity after en bloc tumor removal. Minimal fluorescence remained in the final wound bed, likely from nonenhancing tumor. Fluorescence was heterogeneously distributed at the infiltrative margin in resected tumor pieces imaged ex vivo. Postoperative MRI confirmed gross-total resection. The video can be found here: https://stream.cadmore.media/r10.3171/2021.10.FOCVID21201.

    View details for DOI 10.3171/2021.10.FOCVID21201

    View details for PubMedID 36284595

    View details for PubMedCentralID PMC9557340

  • EGFR-targeted intraoperative fluorescence imaging detects high-grade glioma with panitumumab-IRDye800 in a phase 1 clinical trial. Theranostics Zhou, Q., van den Berg, N. S., Rosenthal, E. L., Iv, M., Zhang, M., Vega Leonel, J. C., Walters, S., Nishio, N., Granucci, M., Raymundo, R., Yi, G., Vogel, H., Cayrol, R., Lee, Y. J., Lu, G., Hom, M., Kang, W., Hayden Gephart, M., Recht, L., Nagpal, S., Thomas, R., Patel, C., Grant, G. A., Li, G. 2021; 11 (15): 7130-7143

    Abstract

    Rationale: First-line therapy for high-grade gliomas (HGGs) includes maximal safe surgical resection. The extent of resection predicts overall survival, but current neuroimaging approaches lack tumor specificity. The epidermal growth factor receptor (EGFR) is a highly expressed HGG biomarker. We evaluated the safety and feasibility of an anti-EGFR antibody, panitumuab-IRDye800, at subtherapeutic doses as an imaging agent for HGG. Methods: Eleven patients with contrast-enhancing HGGs were systemically infused with panitumumab-IRDye800 at a low (50 mg) or high (100 mg) dose 1-5 days before surgery. Near-infrared fluorescence imaging was performed intraoperatively and ex vivo, to identify the optimal tumor-to-background ratio by comparing mean fluorescence intensities of tumor and histologically uninvolved tissue. Fluorescence was correlated with preoperative T1 contrast, tumor size, EGFR expression and other biomarkers. Results: No adverse events were attributed to panitumumab-IRDye800. Tumor fragments as small as 5 mg could be detected ex vivo and detection threshold was dose dependent. In tissue sections, panitumumab-IRDye800 was highly sensitive (95%) and specific (96%) for pathology confirmed tumor containing tissue. Cellular delivery of panitumumab-IRDye800 was correlated to EGFR overexpression and compromised blood-brain barrier in HGG, while normal brain tissue showed minimal fluorescence. Intraoperative fluorescence improved optical contrast in tumor tissue within and beyond the T1 contrast-enhancing margin, with contrast-to-noise ratios of 9.5 ± 2.1 and 3.6 ± 1.1, respectively. Conclusions: Panitumumab-IRDye800 provided excellent tumor contrast and was safe at both doses. Smaller fragments of tumor could be detected at the 100 mg dose and thus more suitable for intraoperative imaging.

    View details for DOI 10.7150/thno.60582

    View details for PubMedID 34158840

    View details for PubMedCentralID PMC8210618

  • Molecular imaging of a fluorescent antibody against epidermal growth factor receptor detects high-grade glioma. Scientific reports Zhou, Q., Vega Leonel, J. C., Santoso, M. R., Wilson, C., van den Berg, N. S., Chan, C. T., Aryal, M., Vogel, H., Cayrol, R., Mandella, M. J., Schonig, F., Lu, G., Gambhir, S. S., Moseley, M. E., Rosenthal, E. L., Grant, G. A. 2021; 11 (1): 5710

    Abstract

    The prognosis for high-grade glioma (HGG) remains dismal and the extent of resection correlates with overall survival and progression free disease. Epidermal growth factor receptor (EGFR) is a biomarker heterogeneously expressed in HGG. We assessed the feasibility of detecting HGG using near-infrared fluorescent antibody targeting EGFR. Mice bearing orthotopic HGG xenografts with modest EGFR expression were imaged in vivo after systemic panitumumab-IRDye800 injection to assess its tumor-specific uptake macroscopically over 14days, and microscopically ex vivo. EGFR immunohistochemical staining of 59 tumor specimens from 35 HGG patients was scored by pathologists and expression levels were compared to that of mouse xenografts. Intratumoral distribution of panitumumab-IRDye800 correlated with near-infrared fluorescence and EGFR expression. Fluorescence distinguished tumor cells with 90% specificity and 82.5% sensitivity. Target-to-background ratios peaked at 14h post panitumumab-IRDye800 infusion, reaching 19.5 in vivo and 7.6 ex vivo, respectively. Equivalent or higher EGFR protein expression compared to the mouse xenografts was present in 77.1% HGG patients. Age, combined with IDH-wildtype cerebral tumor, was predictive of greater EGFR protein expression in human tumors. Tumor specific uptake of panitumumab-IRDye800 provided remarkable contrast and a flexible imaging window for fluorescence-guided identification of HGGs despite modest EGFR expression.

    View details for DOI 10.1038/s41598-021-84831-4

    View details for PubMedID 33707521

  • In vivo photoacoustic tomography of EGFR overexpressed in hepatocellular carcinoma mouse xenograft. Photoacoustics Zhou, Q., Li, Z., Zhou, J., Joshi, B. P., Li, G., Duan, X., Kuick, R., Owens, S. R., Wang, T. D. 2016; 4 (2): 43-54

    Abstract

    EGFR is a promising cell surface target for in vivo imaging that is highly overexpressed in hepatocellular carcinoma (HCC), a common cancer worldwide. Peptides penetrate easily into tumors for deep imaging, and clear rapidly from the circulation to minimize background. We aim to demonstrate use of an EGFR specific peptide to detect HCC xenograft tumors in mice with photoacoustic imaging. Nude mice implanted with human HCC cells that overexpress EGFR were injected intravenously with Cy5.5-labeled EGFR and scrambled control peptides respectively. Photoacoustic images collected from 0 to 24 h. Photoacoustic signal peaked in tumors at 3 h post-injection. Images from 0 to 1.8 cm beneath the skin revealed increased target-to-background (T/B) ratio from tumors. The T/B ratio was significantly greater for the EGFR versus control peptide. Clearance of signal was observed by ∼24 h. EGFR overexpression was validated with immunofluorescence and immunohistochemistry. A peptide specific for EGFR delivered systemically can detect HCC xenograft tumors in vivo with photoacoustic imaging.

    View details for PubMedID 27766208

    View details for PubMedCentralID PMC5066077

  • Flavinated SDHA underlies the change in intrinsic optical properties of oral cancers. Communications biology Marumo, T., Maduka, C. V., Ural, E., Apu, E. H., Chung, S., Tanabe, K., van den Berg, N. S., Zhou, Q., Martin, B. A., Miura, T., Rosenthal, E. L., Shibahara, T., Contag, C. H. 2023; 6 (1): 1134

    Abstract

    The molecular basis of reduced autofluorescence in oral squamous cell carcinoma (OSCC) cells relative to normal cells has been speculated to be due to lower levels of free flavin adenine dinucleotide (FAD). This speculation, along with differences in the intrinsic optical properties of extracellular collagen, lies at the foundation of the design of currently-used clinical optical detection devices. Here, we report that free FAD levels may not account for differences in autofluorescence of OSCC cells, but that the differences relate to FAD as a co-factor for flavination. Autofluorescence from a 70kDa flavoprotein, succinate dehydrogenase A (SDHA), was found to be responsible for changes in optical properties within the FAD spectral region, with lower levels of flavinated SDHA in OSCC cells. Since flavinated SDHA is required for functional complexation with succinate dehydrogenase B (SDHB), decreased SDHB levels were observed in human OSCC tissue relative to normal tissues. Accordingly, the metabolism of OSCC cells was found to be significantly altered relative to normal cells, revealing vulnerabilities for both diagnosis and targeted therapy. Optimizing non-invasive tools based on optical and metabolic signatures of cancers will enable more precise and early diagnosis leading to improved outcomes in patients.

    View details for DOI 10.1038/s42003-023-05510-w

    View details for PubMedID 37945749

  • Flavinated SDHA Underlies the Change in Intrinsic Optical Properties of Oral Cancers. bioRxiv : the preprint server for biology Marumo, T., Maduka, C. V., Ural, E., Apu, E. H., Chung, S., van den Berg, N. S., Zhou, Q., Martin, B. A., Rosenthal, E. L., Shibahara, T., Contag, C. H. 2023

    Abstract

    The molecular basis of reduced autofluorescence in oral squamous cell carcinoma (OSCC) cells relative to normal cells has been speculated to be due to lower levels of free flavin adenine dinucleotide (FAD). This speculation, along with differences in the intrinsic optical properties of extracellular collagen, lie at the foundation of the design of currently-used clinical optical detection devices. Here, we report that free FAD levels may not account for differences in autofluorescence of OSCC cells, but that the differences relate to FAD as a co-factor for flavination. Autofluorescence from a 70 kDa flavoprotein, succinate dehydrogenase A (SDHA), was found to be responsible for changes in optical properties within the FAD spectral region with lower levels of flavinated SDHA in OSCC cells. Since flavinated SDHA is required for functional complexation with succinate dehydrogenase B (SDHB), decreased SDHB levels were observed in human OSCC tissue relative to normal tissues. Accordingly, the metabolism of OSCC cells was found to be significantly altered relative to normal cells, revealing vulnerabilities for both diagnosis and targeted therapy. Optimizing non-invasive tools based on optical and metabolic signatures of cancers will enable more precise and early diagnosis leading to improved outcomes in patients.

    View details for DOI 10.1101/2023.07.30.551184

    View details for PubMedID 37577521

  • Low-count whole-body PET/MRI restoration: an evaluation of dose reduction spectrum and five state-of-the-art artificial intelligence models. European journal of nuclear medicine and molecular imaging Wang, Y. J., Wang, P., Adams, L. C., Sheybani, N. D., Qu, L., Sarrami, A. H., Theruvath, A. J., Gatidis, S., Ho, T., Zhou, Q., Pribnow, A., Thakor, A. S., Rubin, D., Daldrup-Link, H. E. 2023

    Abstract

    To provide a holistic and complete comparison of the five most advanced AI models in the augmentation of low-dose 18F-FDG PET data over the entire dose reduction spectrum.In this multicenter study, five AI models were investigated for restoring low-count whole-body PET/MRI, covering convolutional benchmarks - U-Net, enhanced deep super-resolution network (EDSR), generative adversarial network (GAN) - and the most cutting-edge image reconstruction transformer models in computer vision to date - Swin transformer image restoration network (SwinIR) and EDSR-ViT (vision transformer). The models were evaluated against six groups of count levels representing the simulated 75%, 50%, 25%, 12.5%, 6.25%, and 1% (extremely ultra-low-count) of the clinical standard 3 MBq/kg 18F-FDG dose. The comparisons were performed upon two independent cohorts - (1) a primary cohort from Stanford University and (2) a cross-continental external validation cohort from Tübingen University - in order to ensure the findings are generalizable. A total of 476 original count and simulated low-count whole-body PET/MRI scans were incorporated into this analysis.For low-count PET restoration on the primary cohort, the mean structural similarity index (SSIM) scores for dose 6.25% were 0.898 (95% CI, 0.887-0.910) for EDSR, 0.893 (0.881-0.905) for EDSR-ViT, 0.873 (0.859-0.887) for GAN, 0.885 (0.873-0.898) for U-Net, and 0.910 (0.900-0.920) for SwinIR. In continuation, SwinIR and U-Net's performances were also discreetly evaluated at each simulated radiotracer dose levels. Using the primary Stanford cohort, the mean diagnostic image quality (DIQ; 5-point Likert scale) scores of SwinIR restoration were 5 (SD, 0) for dose 75%, 4.50 (0.535) for dose 50%, 3.75 (0.463) for dose 25%, 3.25 (0.463) for dose 12.5%, 4 (0.926) for dose 6.25%, and 2.5 (0.534) for dose 1%.Compared to low-count PET images, with near-to or nondiagnostic images at higher dose reduction levels (up to 6.25%), both SwinIR and U-Net significantly improve the diagnostic quality of PET images. A radiotracer dose reduction to 1% of the current clinical standard radiotracer dose is out of scope for current AI techniques.

    View details for DOI 10.1007/s00259-022-06097-w

    View details for PubMedID 36633614

  • MRGFUS-DELIVERED FLUORESCENT EGFR/EGFRVIII ANTIBODY ENABLES THERANOSTIC IMAGING OF PEDIATRIC HIGH-GRADE GLIOMA AND PREDICTS RESPONSE TO TARGETED THERAPY Zhou, Q., Aryal, M., Leonel, J., Santoso, M., Zlitni, A., Vogel, H., Cayrol, R., Li, G., Moseley, M. OXFORD UNIV PRESS INC. 2022: 217
  • 89Zr-panitumumab combined with 18F-FDG-PET improves detection and staging of head and neck squamous cell carcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research Lee, Y., van den Berg, N. S., Duan, H., Azevedo, E. C., Ferri, V., Hom, M., Raymundo, R. C., Valencia, A., Castillo, J., Shen, B., Zhou, Q., Freeman, L., Koran, M. E., Kaplan, M. J., Colevas, A. D., Baik, F. M., Chin, F. T., Martin, B. A., Iagaru, A., Rosenthal, E. L. 2022

    Abstract

    PURPOSE: Determine the safety and specificity of a tumor-targeted radiotracer (89Zr-pan) in combination with 18F-FDG PET/CT to improve diagnostic accuracy in head and neck squamous cell carcinoma (HNSCC).EXPERIMENTAL DESIGN: Adult patients with biopsy-proven HNSCC scheduled for standard of care surgery were enrolled in a clinical trial and underwent systemic administration of 89Zirconium-panitumumab and panitumumab-IRDye800 followed by preoperative 89Zr-pan PET/CT and intraoperative fluorescence imaging. The sensitivity, specificity, and AUC were evaluated.RESULTS: A total of fourteen patients were enrolled and completed the study. Four patients (28.5%) had areas of high 18F-FDG uptake outside the head and neck region with maximum standardized uptake values (SUVmax) greater than 2.0 that were not detected on 89Zr-pan PET/CT. These four patients with incidental findings underwent further workup and had no evidence of cancer on biopsy or clinical follow-up. Forty-eight lesions (primary tumor, LNs, incidental findings) with SUVmax ranging 2.0 - 23.6 were visualized on 18F-FDG PET/CT; 34 lesions on 89Zr-pan PET/CT with SUVmax ranging 0.9 - 10.5. The combined ability of 18F-FDG PET/CT and 89Zr-pan PET/CT to detect HNSCC in the whole body was improved with higher specificity of 96.3% (confidence interval (CI) 89.2 - 100%) compared to 18F-FDG PET/CT alone with specificity of 74.1% (CI 74.1 - 90.6%). One possibly related grade 1 adverse event of prolonged QTc (460 ms) was reported but resolved in follow-up.CONCLUSIONS: 89Zr-pan PET/CT imaging is safe and may be valuable in discriminating incidental findings identified on 18F-FDG-PET/CT from true positive lesions and in localizing metastatic LNs.

    View details for DOI 10.1158/1078-0432.CCR-22-0094

    View details for PubMedID 35929985

  • The Use of Panitumumab-IRDye800CW in a Novel Murine Model for Conjunctival Squamous Cell Carcinoma. Translational vision science & technology Youn, G. M., Case, A. G., Jarin, T., Li, B., Swarup, A., Naranjo, A., Bou-Khalil, C., Yao, J., Zhou, Q., Hom, M. E., Rosenthal, E. L., Wu, A. Y. 2022; 11 (7): 23

    Abstract

    Purpose: Conjunctival squamous cell carcinoma (SCC) is a sight-threatening ocular surface malignancy with the primary treatment modality being surgical resection. To evaluate surgical imaging modalities to improve surgical resection, we established a novel murine model for conjunctival SCC to demonstrate the utility of panitumumab-IRDye800, a fluorescently labeled anti-epidermal growth factor receptor (EGFR) antibody.Methods: NOD-scid IL2Rgammanull (NSG) mice received subconjunctival injection of UM-SCC-1 or SCC-9, head and neck SCC cell lines. On tumor growth, mice were injected with Panitumumab-IRDye800CW, and imaged with a small animal imaging system and optical coherence tomography (OCT). Immunohistochemistry for SCC markers were used to confirm tumor origin.Results: Seventy-five percent (N = 4) of the UM-SCC-1 group developed aggressive, rapidly growing tumors that were P40 and EGFR positive within two weeks of inoculation. The SCC-9 tumors failed to demonstrate any growth (N = 4). Ocular tumors demonstrated high fluorescence levels with a tumor to background ratio of 3.8.Conclusions: Subconjunctival injections are an appropriate technique to create in vivo models for assessing treatment modalities and novel therapies in conjunctival SCC.Translational Relevance: This model demonstrates Panitumumab-IRDye800CW's utility in the ophthalmic setting and suggests that clinical trials may be warranted.

    View details for DOI 10.1167/tvst.11.7.23

    View details for PubMedID 35895055

  • Noninvasive ultrasonic induction of cerebrospinal fluid flow enhances intrathecal drug delivery. Journal of controlled release : official journal of the Controlled Release Society Aryal, M., Azadian, M. M., Hart, A. R., Macedo, N., Zhou, Q., Rosenthal, E. L., Airan, R. D. 2022

    Abstract

    Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.

    View details for DOI 10.1016/j.jconrel.2022.06.067

    View details for PubMedID 35798095

  • MRI Radiogenomics of Pediatric Medulloblastoma: A Multicenter Study. Radiology Zhang, M., Wong, S. W., Wright, J. N., Wagner, M. W., Toescu, S., Han, M., Tam, L. T., Zhou, Q., Ahmadian, S. S., Shpanskaya, K., Lummus, S., Lai, H., Eghbal, A., Radmanesh, A., Nemelka, J., Harward, S. 2., Malinzak, M., Laughlin, S., Perreault, S., Braun, K. R., Lober, R. M., Cho, Y. J., Ertl-Wagner, B., Ho, C. Y., Mankad, K., Vogel, H., Cheshier, S. H., Jacques, T. S., Aquilina, K., Fisher, P. G., Taylor, M., Poussaint, T., Vitanza, N. A., Grant, G. A., Pfister, S., Thompson, E., Jaju, A., Ramaswamy, V., Yeom, K. W. 2022: 212137

    Abstract

    Background Radiogenomics of pediatric medulloblastoma (MB) offers an opportunity for MB risk stratification, which may aid therapeutic decision making, family counseling, and selection of patient groups suitable for targeted genetic analysis. Purpose To develop machine learning strategies that identify the four clinically significant MB molecular subgroups. Materials and Methods In this retrospective study, consecutive pediatric patients with newly diagnosed MB at MRI at 12 international pediatric sites between July 1997 and May 2020 were identified. There were 1800 features extracted from T2- and contrast-enhanced T1-weighted preoperative MRI scans. A two-stage sequential classifier was designed-one that first identifies non-wingless (WNT) and non-sonic hedgehog (SHH) MB and then differentiates therapeutically relevant WNT from SHH. Further, a classifier that distinguishes high-risk group 3 from group 4 MB was developed. An independent, binary subgroup analysis was conducted to uncover radiomics features unique to infantile versus childhood SHH subgroups. The best-performing models from six candidate classifiers were selected, and performance was measured on holdout test sets. CIs were obtained by bootstrapping the test sets for 2000 random samples. Model accuracy score was compared with the no-information rate using the Wald test. Results The study cohort comprised 263 patients (mean age ± SD at diagnosis, 87 months ± 60; 166 boys). A two-stage classifier outperformed a single-stage multiclass classifier. The combined, sequential classifier achieved a microaveraged F1 score of 88% and a binary F1 score of 95% specifically for WNT. A group 3 versus group 4 classifier achieved an area under the receiver operating characteristic curve of 98%. Of the Image Biomarker Standardization Initiative features, texture and first-order intensity features were most contributory across the molecular subgroups. Conclusion An MRI-based machine learning decision path allowed identification of the four clinically relevant molecular pediatric medulloblastoma subgroups. © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Verschakelen in this issue.

    View details for DOI 10.1148/radiol.212137

    View details for PubMedID 35438562

  • Fluorescent molecular imaging can improve intraoperative sentinel margin detection in oral squamous cell carcinoma. Journal of nuclear medicine : official publication, Society of Nuclear Medicine Krishnan, G., van den Berg, N. S., Nishio, N., Kapoor, S., Pei, J., Freeman, L., Lee, Y., van Keulen, S., Fakurnejad, S., Condon, J., Back, F., Martin, B., Rosenthal, E. L. 1800

    Abstract

    Rationale: In head and neck cancer, a major limitation of current intraoperative margin analysis is the ability to detect areas most likely to be positive based on specimen palpation, especially for larger specimens where sampling error limits detection of positive margins. This study aims to prospectively examine the clinical value of fluorescent molecular imaging to accurately identify "the sentinel margin," the point on a specimen where tumour lies closest to the resected edge in real-time during frozen section analysis. Methods: Eighteen patients with oral squamous cell carcinoma were enrolled into a prospective clinical trial and infused intravenously with 50mg of panitumumab-IRDye800CW 1-5 days prior to surgery. Resected specimens were imaged in a closed-field near-infrared optical imaging system in near-real time, and custom designed software was used to identify locations of highest fluorescence on deep and peripheral margins. The surgeon identified the sentinel margin blinded to optical specimen mapping, and then the regions of highest fluorescence were identified and marked for frozen analysis. Final pathology based on specimen reconstruction was used as reference standard. Results: Resected specimens were imaged in the operating room and fluorescence had a higher interobserver agreement with pathology (Cohen kappa value 0.96) than the surgeon (Cohen kappa values of 0.82) for the location of the closest margin. Plotting margin distance at the predicted sentinel margin location of each observer versus the actual closest margin distance at pathology demonstrated best correlation between fluorescence and pathology (R2 = 0.98), with surgeon (R2 = 0.75). Principal Conclusion: Fluorescence imaging can improve identification of the sentinel margin in head and neck cancer resections, holding promise for rapid identification of positive margins and improved oncological outcomes.

    View details for DOI 10.2967/jnumed.121.262235

    View details for PubMedID 35027369

  • EGFR-targeted fluorescence-guided surgery: differential imaging performance and clinical predictors in solid tumors World Molecular Imaging Congress (WMIC) Zhou, Q., Iv, M., Moseley, M. 2022
  • Metastatic and sentinel lymph node mapping using intravenously delivered Panitumumab-IRDye800CW. Theranostics Krishnan, G., van den Berg, N. S., Nishio, N., Juniper, G., Pei, J., Zhou, Q., Lu, G., Lee, Y. J., Ramos, K., Iagaru, A. H., Baik, F. M., Colevas, A. D., Martin, B. A., Rosenthal, E. L. 2021; 11 (15): 7188-7198

    Abstract

    Rationale: Sentinel lymph node biopsy (SLNB) is a well-established minimally invasive staging procedure that maps the spread of tumour metastases from their primary site to the regional lymphatics. Currently, the procedure requires the local peri-tumoural injection of radiolabelled and/or optical agents, and is therefore operator dependent, disruptive to surgical workflow and restricted largely to a small subset of malignancies that can be readily accessed externally for local tracer injection. The present study set out to determine whether intravenous (IV) infusion of a tumor-targeted tracer could identify sentinel and metastatic lymph nodes (LNs) in order to overcome these limitations. Methods: We examined 27 patients with oral squamous cell carcinoma (OSCC), 18 of whom were clinically node negative (cN0). Patients were infused intravenously with 50mg of Panitumumab-IRDye800CW prior to surgical resection of their primary tumour with neck dissection and/or SLNB. Lymphadenectomy specimens underwent fluorescence molecular imaging to evaluate tracer distribution to LNs. Results: A total of 960 LNs were analysed, of which 34 (3.5%) contained metastatic disease. Panitumumab-IRDye800CW preferentially localized to metastatic and sentinel LNs as evidenced by a higher fluorescent signal relative to other lymph nodes. The median MFI of metastatic LNs was significantly higher than the median MFI of benign LNs (0.06 versus 0.02, p < 0.05). Furthermore, selecting the highest five fluorescence intensity LNs from individual specimens resulted in 100% sensitivity, 85.8% specificity and 100% negative predictive value (NPV) for the detection of occult metastases and 100% accuracy for clinically staging the neck. In the cN+ cohort, assessment of the highest 5 fluorescence LNs per patient had 87.5% sensitivity, 93.2% specificity and 99.1% NPV for the detection of metastatic nodes. Conclusion: When intravenously infused, a tumour-targeted tracer localized to sentinel and metastatic lymph nodes. Further validation of an IV tumor-targeted tracer delivery approach for SLNB could dramatically change the practice of SLNB, allowing its application to other malignancies where the primary tumour is not accessible for local tracer injection.

    View details for DOI 10.7150/thno.55389

    View details for PubMedID 34158844

    View details for PubMedCentralID PMC8210603

  • In Vivo Evaluation of Near-Infrared Fluorescent Probe for TIM3 Targeting in Mouse Glioma. Molecular imaging and biology Zhang, M., Zhou, Q., Huang, C., Chan, C. T., Wu, W., Li, G., Lim, M., Gambhir, S. S., Daldrup-Link, H. E. 2021

    Abstract

    Current checkpoint inhibitor immunotherapy strategies in glioblastoma are challenged by mechanisms of resistance including an immunosuppressive tumor microenvironment. T cell immunoglobulin domain and mucin domain 3 (TIM3) is a late-phase checkpoint receptor traditionally associated with T cell exhaustion. We apply fluorescent imaging techniques to explore feasibility of in vivo visualization of the immune state in a glioblastoma mouse model.TIM3 monoclonal antibody was conjugated to a near-infrared fluorescent dye, IRDye-800CW (800CW). The TIM3 experimental conjugate and isotype control were assessed for specificity with immunofluorescent staining and flow cytometry in murine cell lines (GL261 glioma and RAW264.7 macrophages). C57BL/6 mice with orthotopically implanted GL261 cells were imaged in vivo over 4 days after intravenous TIM3-800CW injection to assess tumor-specific uptake. Cell-specific uptake was then assessed on histologic sections.The experimental TIM3-800CW, but not its isotype control, bound to RAW264.7 macrophages in vitro. Specificity to RAW264.7 macrophages and not GL261 tumor cells was quantitatively confirmed with the corresponding clone of TIM3 on flow cytometry. In vivo fluorescence imaging of the 800CW signal was localized to the intracranial tumor and significantly higher for the TIM3-800CW cohort, relative to non-targeting isotype control, immediately after tail vein injection and for up to 48 h after injection. Resected organs of tumor bearing mice showed significantly higher uptake in the liver and spleen. TIM3-800CW was seen to co-stain with CD3 (13%), CD11b (29%), and CD206 (26%).We propose fluorescent imaging of immune cell imaging as a potential strategy for monitoring and localizing immunologically relevant foci in the setting of brain tumors. Alternative markers and target validation will further clarify the temporal relationship of immunosuppressive effector cells throughout glioma resistance.

    View details for DOI 10.1007/s11307-021-01667-0

    View details for PubMedID 34846678

  • Focused ultrasound: growth potential and future directions in neurosurgery. Journal of neuro-oncology Zhang, M., Rodrigues, A., Zhou, Q., Li, G. 2021

    Abstract

    Over the past two decades, vast improvements in focused ultrasound (FUS) technology have made the therapy an exciting addition to the neurosurgical armamentarium. In this time period, FUS has gained US Food and Drug Administration (FDA) approval for the treatment of two neurological disorders, and ongoing efforts seek to expand the lesion profile that is amenable to ultrasonic intervention. In the following review, we highlight future applications for FUS therapy and compare its potential role against established technologies, including deep brain stimulation and stereotactic radiosurgery. Particular attention is paid to tissue ablation, blood-brain-barrier opening, and gene therapy. We also address technical and infrastructural challenges involved with FUS use and summarize the hurdles that must be overcome before FUS becomes widely accepted in the neurosurgical community.

    View details for DOI 10.1007/s11060-021-03820-9

    View details for PubMedID 34410576

  • FIRST-IN-HUMAN FLUORESCENCE GUIDED SURGERY OF HIGH-GRADE GLIOMAS USING PANITUMUMAB-IRDYE800 Zhou, Q., van den Berg, N., Nishio, N., Lu, G., Chirita, S., Raymundo, R., Yi, G., Vogel, H., Cayrol, R., Rosenthal, E., Li, G. OXFORD UNIV PRESS INC. 2020: 52
  • Effect of Formalin Fixation for Near-Infrared Fluorescence Imaging with an Antibody-Dye Conjugate in Head and Neck Cancer Patients. Molecular imaging and biology Kapoor, S., Lu, G., van den Berg, N. S., Krishnan, G., Pei, J., Zhou, Q., Martin, B. A., Baik, F. M., Rosenthal, E. L., Nishio, N. 2020

    Abstract

    PURPOSE: This study evaluated the effect of formalin fixation for near-infrared (NIR) fluorescence imaging of an antibody-dye complex (panitumumab-IRDye800CW) that was intravenously administered to patients with head and neck squamous cell carcinoma (HNSCC) scheduled to undergo surgery of curative intent.PROCEDURES: HNSCC patients were infused with 25 or 50mg of panitumumab-IRDye800CW followed by surgery 1-5days later. Following resection, primary tumor specimens were imaged in a closed-field fluorescence imaging device, before and after formalin fixation. The fluorescence images of formalin-fixed specimens were compared with images prior to formalin fixation. Regions of interest were drawn on the primary tumor and on the adjacent normal tissue on the fluorescence images. The mean fluorescence intensity (MFI) and tumor-to-background ratios (TBRs) of the fresh and formalin-fixed tissues were compared.RESULTS: Of the 30 enrolled patients, 20 tissue specimens were eligible for this study. Formalin fixation led to an average of 10% shrinkage in tumor specimen size (p<0.0001). Tumor MFI in formalin-fixed specimens was on average 10.9% lower than that in the fresh specimens (p=0.0002). However, no statistical difference was found between the TBRs of the fresh specimens and those of the formalin-fixed specimens (p=0.85).CONCLUSIONS: Despite the 11% decrease in MFI between fresh and formalin-fixed tissue specimens, the relative difference between tumor and normal tissue as measured in TBR remained unchanged. This data suggests that evaluation of formalin-fixed tissue for assessing the accuracy of fluorescence-guided surgery approaches could provide a valid, yet more flexible, alternative to fresh tissue analysis.TRIAL REGISTRATION: NCT02415881.

    View details for DOI 10.1007/s11307-020-01553-1

    View details for PubMedID 33078373

  • Tumour-specific fluorescence-guided surgery for pancreatic cancer using panitumumab-IRDye800CW: a phase 1 single-centre, open-label, single-arm, dose-escalation study. The lancet. Gastroenterology & hepatology Lu, G., van den Berg, N. S., Martin, B. A., Nishio, N., Hart, Z. P., van Keulen, S., Fakurnejad, S., Chirita, S. U., Raymundo, R. C., Yi, G., Zhou, Q., Fisher, G. A., Rosenthal, E. L., Poultsides, G. A. 2020

    Abstract

    BACKGROUND: Complete surgical resection remains the primary curative option for pancreatic ductal adenocarcinoma, with positive margins in 30-70% of patients. In this study, we aimed to evaluate the use of intraoperative tumour-specific imaging to enhance a surgeon's ability to detect visually occult cancer in real time.METHODS: In this single-centre, open-label, single-arm study, done in the USA, we enrolled patients who had clinically suspicious or biopsy-confirmed pancreatic ductal adenocarcinomas and were scheduled for curative surgery. Eligible patients were 19 years of age or older with a life expectancy of more than 12 weeks and a Karnofsky performance status of at least 70% or an Eastern Cooperative Oncology Group or Zubrod level of one or lower, who were scheduled to undergo curative surgery. Patients were sequentially enrolled into each dosing group and 2-5 days before surgery, patients were intravenously infused with 100 mg of unlabelled panitumumab followed by 25 mg, 50 mg, or 75 mg of the near-infrared fluorescently labelled antibody (panitumumab-IRDye800CW). The primary endpoint was to determine the optimal dose of panitumumab-IRDye800CW in identifying pancreatic ductal adenocarcinomas as measured by tumour-to-background ratio in all patients. The tumour-to-background ratio was defined as the fluorescence signal of the tumour divided by the fluorescence signal of the surrounding healthy tissue. The dose-finding part of this study has been completed. This study is registered with ClinicalTrials.gov, NCT03384238.FINDINGS: Between April, 2018, and July, 2019, 16 patients were screened for enrolment onto the study. Of the 16 screened patients, two (12%) patients withdrew from the study and three (19%) were not eligible; 11 (69%) patients completed the trial, all of whom were clinically diagnosed with pancreatic ductal adenocarcinoma. The mean tumour-to-background ratio of primary tumours was 3·0 (SD 0·5) in the 25 mg group, 4·0 (SD 0·6) in the 50 mg group, and 3·7 (SD 0·4) in the 75 mg group; the optimal dose was identified as 50 mg. Intraoperatively, near-infrared fluorescence imaging provided enhanced visualisation of the primary tumours, metastatic lymph nodes, and small (<2 mm) peritoneal metastasis. Intravenous administration of panitumumab-IRDye800CW at the doses of 25 mg, 50 mg, and 75 mg did not result in any grade 3 or higher adverse events. There were no serious adverse events attributed to panitumumab-IRDye800CW, although four possibly related adverse events (grade 1 and 2) were reported in four patients.INTERPRETATION: To our knowledge, this study presents the first clinical use of panitumumab-IRDye800CW for detecting pancreatic ductal adenocarcinomas and shows that panitumumab-IRDye800CW is safe and feasible to use during pancreatic cancer surgery. Tumour-specific intraoperative imaging might have added value for treatment of patients with pancreatic ductal adenocarcinomas through improved patient selection and enhanced visualisation of surgical margins, metastatic lymph nodes, and distant metastasis.FUNDING: National Institutes of Health and the Netherlands Organization for Scientific Research.

    View details for DOI 10.1016/S2468-1253(20)30088-1

    View details for PubMedID 32416764

  • Optimal Dosing Strategy for Fluorescence-Guided Surgery with Panitumumab-IRDye800CW in Head and Neck Cancer MOLECULAR IMAGING AND BIOLOGY Nishio, N., van den Berg, N. S., van Keulen, S., Martin, B. A., Fakurnejad, S., Zhou, Q., Lu, G., Chirita, S. U., Kaplan, M. J., Divi, V., Colevas, A. D., Rosenthal, E. L. 2020; 22 (1): 156–64
  • Predicting Therapeutic Antibody Delivery into Human Head and Neck Cancers. Clinical cancer research : an official journal of the American Association for Cancer Research Lu, G., Fakurnejad, S., Martin, B. A., van den Berg, N. S., van Keulen, S., Nishio, N., Zhu, A. J., Chirita, S. U., Zhou, Q., Gao, R. W., Kong, C. S., Fischbein, N., Penta, M., Colevas, A. D., Rosenthal, E. L. 2020

    Abstract

    PURPOSE: The efficacy of antibody-based therapeutics depends on successful drug delivery into solid tumors, therefore there is a clinical need to measure intratumoral antibody distribution. This study aims to develop and validate an imaging and computation platform to directly quantify and predict antibody delivery into human head and neck cancers in a clinical study.EXPERIMENTAL DESIGN: Twenty-four patients received systemic infusion of a near-infrared (NIR) fluorescence-labeled therapeutic antibody followed by surgical tumor resection. A computational platform was developed to quantify the extent of heterogeneity of intratumoral antibody distribution. Both univariate and multivariate regression analysis were used to select the most predictive tumor biological factors for antibody delivery. Quantitative image features from the pre-treatment magnetic resonance imaging (MRI) were extracted and correlated with fluorescence imaging of antibody delivery.RESULTS: This study not only confirmed heterogeneous intratumoral antibody distribution in line with many preclinical reports, but also quantified the extent of inter-patient, inter-tumor, and intra-tumor heterogeneity of antibody delivery. This study demonstrated the strong predictive value of tumor size for intratumoral antibody accumulation and its significant impact on antibody distribution in both primary tumor and lymph node metastasis. Furthermore, this study established the feasibility of using contrast-enhanced MRI to predict antibody delivery.CONCLUSIONS: This study provides a clinically translatable platform to measure antibody delivery into solid tumors and yields valuable insight into clinically relevant antibody tumor penetration, with implications in the selection of patients amenable to antibody therapy and the design of more effective dosing strategies.

    View details for DOI 10.1158/1078-0432.CCR-19-3717

    View details for PubMedID 31980465

  • Safety and Stability of Antibody-Dye Conjugate in Optical Molecular Imaging. Molecular imaging and biology Pei, J. n., Juniper, G. n., van den Berg, N. S., Nisho, N. n., Broadt, T. n., Welch, A. R., Yi, G. S., Raymundo, R. C., Chirita, S. U., Lu, G. n., Krishnan, G. n., Lee, Y. J., Kapoor, S. n., Zhou, Q. n., Colevas, A. D., Lui, N. S., Poultsides, G. A., Li, G. n., Zinn, K. R., Rosenthal, E. L. 2020

    Abstract

    The development of molecularly targeted tracers is likely to improve the accuracy of diagnostic, screening, and therapeutic tools. Despite the many therapeutic antibodies that are FDA-approved with known toxicity, only a limited number of antibody-dye conjugates have been introduced to the clinic. Thorough evaluation of the safety, stability, and pharmacokinetics of antibody conjugates in the clinical setting compared with their parental components could accelerate the clinical approval of antibodies as agents for molecular imaging. Here we investigate the safety and stability of a near-infrared fluorescent dye (IRDye800CW) conjugated panitumumab, an approved therapeutic antibody, and report on the product stability, pharmacokinetics, adverse events, and QTc interval changes in patients.Panitumumab-IRDye800CW was made under good manufacturing practice (GMP) conditions in a single batch on March 26, 2014, and then evaluated over 4.5 years at 0, 3, and 6 months, and then at 6-month intervals thereafter. We conducted early phase trials in head and neck, lung, pancreas, and brain cancers with panitumumab-IRDye800CW. Eighty-one patients scheduled to undergo standard-of-care surgery were infused with doses between 0.06 to 2.83 mg/kg of antibody. Patient ECGs, blood samples, and adverse events were collected over 30-day post-infusion for analysis.Eighty-one patients underwent infusion of the study drug at a range of doses. Six patients (7.4 %) experienced an adverse event that was considered potentially related to the drug. The most common event was a prolonged QTc interval which occurred in three patients (3.7 %). Panitumumab-IRDye800CW had two OOS results at 42 and 54 months while meeting all other stability testing criteria.Panitumumab-IRDye800CW was safe and stable to administer over a 54-month window with a low rate of adverse events (7.4 %) which is consistent with the rate associated with panitumumab alone. This data supports re-purposing therapeutic antibodies as diagnostic imaging agents with limited preclinical toxicology studies.

    View details for DOI 10.1007/s11307-020-01536-2

    View details for PubMedID 32880818

  • Endoscopic Fluorescence-Guided Surgery for Sinonasal Cancer Using an Antibody-Dye Conjugate. The Laryngoscope Hart, Z. P., Nishio, N., Krishnan, G., Lu, G., Zhou, Q., Fakurnejad, S., Wormald, P. J., van den Berg, N. S., Rosenthal, E. L., Baik, F. M. 2019

    Abstract

    OBJECTIVE: Endoscopic resection of sinonasal squamous cell carcinoma has become the standard of care, but challenges remain in obtaining clear resection margins. The current study evaluated the feasibility of endoscopic fluorescence-guided surgery (FGS) to improve surgical resection in a human sinus surgical model.METHODS: A fluorescence endoscope optimized for near-infrared (NIR) fluorescence detection was evaluated in a phantom study. Various endoscope diameters (4 and 10mm) and viewing angles (0, 30, and 45 degrees) were evaluated to determine the sensitivity of the system for IRDye800CW detection at various working distances (1-5 cm). Endoscopic FGS was then validated in a three-dimensional human sinus surgical model to which squamous cell tumors derived from mice were inserted. Mice had received intravenous panitumumab-IRDye800CW and upon fluorescence-guided tumor resection, mean fluorescence intensity (MFI) and tumor-to-background ratio (TBR) were calculated in in situ and ex vivo settings.RESULTS: A significantly higher fluorescence intensity was found when using the 10-mm diameter endoscope compared to the 4mm diameter endoscope (P<.001). No significant difference in MFI was found among the viewing angles of the 4-mm diameter endoscope. Using the human sinus model, the highest MFI and TBR were obtained at a 1-cm working distance compared to longer working distances.CONCLUSION: We demonstrate that clinically acceptable TBRs were obtained with several working distances to discriminate tumor tissue from adjacent normal tissue in a human sinus model, and that endoscopic FGS may have great potential in identifying residual tumor tissue regions during surgery. Laryngoscope, 2019.

    View details for DOI 10.1002/lary.28483

    View details for PubMedID 31854462

  • Probe-based fluorescence dosimetry of an antibody-dye conjugate to identify head and neck cancer as a first step to fluorescence-guided tissue preselection for pathological assessment. Head & neck Nishio, N., van Keulen, S., van den Berg, N. S., Lu, G., LaRochelle, E. P., Davis, S. C., Martin, B. A., Fakurnejad, S., Zhou, Q., Birkeland, A. C., Kaplan, M. J., Divi, V., Colevas, A. D., Pogue, B. W., Rosenthal, E. L. 2019

    Abstract

    BACKGROUND: Despite the rapid growth of fluorescence imaging, accurate sampling of tissue sections remains challenging. Development of novel technologies to improve intraoperative assessment of tissue is needed.METHODS: A novel contact probe-based fluorescence dosimeter device, optimized for IRDye800CW quantification, was developed. After evaluation of the device in a phantom setup, its clinical value was defined ex vivo in patients with head and neck squamous cell carcinoma who received panitumumab-IRDye800CW.RESULTS: Ten patients were enrolled with a total of 216 data points obtained. Final histopathology showed tumor in 119 spots and normal tissue in 97 spots. Fluorescence-to-excitation ratios in tumor tissue were more than three times higher than those in normal tissue. The area under the curve was 0.86 (95% CI: 0.81-0.91) for tumor detection.CONCLUSIONS: Fluorescence-guided tissue preselection using a fluorescence dosimeter could have substantial impact on tissue sampling for frozen section analysis and potentially reduce sampling errors.

    View details for DOI 10.1002/hed.25964

    View details for PubMedID 31571335

  • The Clinical Application of Fluorescence-Guided Surgery in Head and Neck Cancer JOURNAL OF NUCLEAR MEDICINE van Keulen, S., Nishio, N., Fakurnejad, S., Birkeland, A., Martin, B. A., Lu, G., Zhou, Q., Chirita, S. U., Forouzanfar, T., Colevas, A., van den Berg, N. S., Rosenthal, E. L. 2019; 60 (6): 758–63
  • Optimal Dosing Strategy for Fluorescence-Guided Surgery with Panitumumab-IRDye800CW in Head and Neck Cancer. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging Nishio, N., van den Berg, N. S., van Keulen, S., Martin, B. A., Fakurnejad, S., Zhou, Q., Lu, G., Chirita, S. U., Kaplan, M. J., Divi, V., Colevas, A. D., Rosenthal, E. L. 2019

    Abstract

    PURPOSE: To identify the optimal dosing strategy for fluorescence-guided surgery in patients with head and neck squamous cell carcinoma, we conducted a dose-ranging study evaluating the anti-epidermal growth factor receptor (EGFR) therapeutic antibody, panitumumab, that was fluorescently labeled with the near-infrared dye IRDye800CW.PROCEDURES: Patients (n=24) received either 0.5 or 1.0mg/kg panitumumab-IRDye800CW in the weight-based dosing group or 25 or 50mg panitumumab-IRDye800CW in the fixed dosing group. Following surgery, whole primary specimens were imaged in a closed-field device and the mean fluorescence intensity (MFI) and tumor-to-background ratio (TBR) were assessed. Clinical variables, including dose, time of infusion-to-surgery, age, unlabeled dose, gender, primary tumor site, and tumor size, were analyzed to evaluate the factors affecting the fluorescence intensity in order to identify the optimal dose for intraoperative fluorescence imaging.RESULTS: A total of 24 primary tumor specimens were imaged and analyzed in this study. Although no correlations between TBR and dose of panitumumab-IRDye800CW were found, there were moderate-strong correlations between the primary tumor MFI and panitumumab-IRDye800CW dose for fixed dose (mg) (R2=0.42) and for dose/weight (mg/kg) (R2=0.54). Results indicated that the optimal MFI was at approximately 50mg for fixed dose and 0.75mg/kg for dose/weight. No significant differences were found for the primary tumor MFI and TBRs between the weight-based dosing and the fixed dosing groups. MFIs significantly increased when the infusion-to-surgery window was reduced to within 2days (vs. 3days or more, p<0.05).CONCLUSIONS: Antibody-based imaging for surgical resection is under investigation in multiple clinical trials. Our data suggests that a fixed dose of 50mg is an appropriate diagnostic dose for successful surgical fluorescence imaging.

    View details for PubMedID 31054001

  • The Clinical Application of Fluorescence-Guided Surgery in Head and Neck Cancer. Journal of nuclear medicine : official publication, Society of Nuclear Medicine van Keulen, S., Nishio, N., Fakurnejad, S., Birkeland, A., Martin, B. A., Lu, G., Zhou, Q., Chirita, S. U., Forouzanfar, T., Colevas, D., van den Berg, N. S., Rosenthal, E. L. 2019

    Abstract

    Although surgical resection has been the primary treatment modality of solid tumors for decades, surgeons still rely on visual cues and palpation to delineate healthy from cancerous tissue. This may contribute to the high rate (up to 30%) of positive margins in head and neck cancer resections. Margin status in these patients is the most important prognostic factor for overall survival. In addition, second primary lesions may be present at the time of surgery. Although often unnoticed by the medical team, these lesions can have significant survival ramifications. We hypothesize that real-time fluorescence imaging can enhance intraoperative decision-making by aiding the surgeon in detecting close or positive margins and visualizing unanticipated regions of primary disease. The purpose of this study was to assess the clinical utility of real-time fluorescence imaging for intraoperative decision-making. Methods: Head and neck cancer patients (n = 14) scheduled for curative resection were enrolled in a clinical trial evaluating panitumumab-IRDye800CW for surgical guidance (NCT02415881). Open-field fluorescence imaging was performed throughout the surgical procedure. The fluorescence signal was quantified as signal-to-background ratios to characterize the fluorescence contrast of regions of interest relative to background. Results: Fluorescence imaging was able to improve surgical decision-making in three cases (21.4%); identification of a close margin (n = 1) and unanticipated regions of primary disease (n = 2). Conclusion: This study demonstrates the clinical applications of fluorescence imaging on intraoperative decision-making. This information is required for designing phase III clinical trials using this technique. Furthermore, this study is the first to demonstrate this application for intraoperative decision-making during resection of primary tumors.

    View details for PubMedID 30733319

  • Rapid, non-invasive fluorescence margin assessment: Optical specimen mapping in oral squamous cell carcinoma. Oral oncology van Keulen, S., van den Berg, N. S., Nishio, N., Birkeland, A., Zhou, Q., Lu, G., Wang, H., Middendorf, L., Forouzanfar, T., Martin, B. A., Colevas, A. D., Rosenthal, E. L. 2019; 88: 58–65

    Abstract

    OBJECTIVE: Surgical resection remains the primary treatment for the majority of solid tumors. Despite efforts to obtain wide margins, close or positive surgical margins (<5 mm) are found in 15-30% of head and neck cancer patients. Obtaining negative margins requires immediate, intraoperative feedback of margin status. To this end, we propose optical specimen mapping of resected tumor specimens immediately after removal.MATERIALS AND METHODS: A first-in-human pilot study was performed in patients (n = 8) after infusion of fluorescently labeled antibody, panitumumab-IRDye800 to allow surgical mapping of the tumor specimen. Patients underwent standard of care surgical resection for head and neck squamous cell carcinoma (HNSCC). Optical specimen mapping was performed on the primary tumor specimen and correlated with pathological findings after tissue processing.RESULTS: Optical mapping of the specimen had a 95% sensitivity and 89% specificity to detect cancer within 5 mm (n = 160) of the cut surface. To detect tumor within 2 mm of the specimen surface, the sensitivity of optical specimen mapping was 100%. The maximal observed penetration depth of panitumumab-IRDye800 through human tissue in our study was 6.3 mm.CONCLUSION: Optical specimen mapping is a highly sensitive and specific method for evaluation of margins within <5 mm of the tumor mass in HNSCC specimens. This technology has potentially broad applications for ensuring adequate tumor resection and negative margins in head and neck cancers.

    View details for PubMedID 30616798

  • Predicting antibody penetration in a first-in-human clinical trial of head and neck cancers Society of Photographic Instrumentation Engineers (SPIE) Photonics West Lu, G., Fakurnejad, S., Martin, B. A., Zhu, A., van den Berg, N., van Keulen, S., Zhou, Q., Teraphongphom , N., Gao, R., Oberhelman, N., Chirita, S. U., Erstey, R., Kong , C., Colevas, A., Rosenthal, E. 2019: 1085905
  • Fluorescence-guided surgery with panitumumab-IRDye800 and cetuximab-IRDye800 in glioblastoma patients Society of Photographic Instrumentation Engineers (SPIE) Photonics West Zhou, Q., Miller, S. E., van den Berg, N., Lu, G., Vogel, H., Cayrol, R., Oberhelman , N. J., Chirita , S. U., van Keulen, S., Grant, G. A., Li, G., Rosenthal, E. 2019: 108620Y
  • Reversible blood-brain barrier modulation enhances in vivo delivery of panitumumab-IRDye800 to high-grade glioma in cranial window model Society of Photographic Instrumentation Engineers (SPIE) Photonics West Zhou, Q., Wilson, C. M., Vogel, H., Teraphongphom , N., Ertsey, R. D., Chu, P. M., Lu, G., van den Berg, N., van Keulen, S., Nishio, N., Grant, G. A. 2019 : 1086402
  • Clinical Application of Fluorescence Guided Surgery in Head and Neck Cancer: Successes and Limitations Triological Society 122nd Annual Meeting at Combined Otolaryngology Spring Meetings (COSM) van Keulen, S., van den Berg, N., Nishio, N., Lu, G., Zhou, Q., Rosenthal, E. 2019: 25
  • Rapid, non-invasive fluorescence margin assessment: Optical specimen mapping in oral squamous cell carcinoma ORAL ONCOLOGY van Keulen, S., van den Berg, N. S., Nishio, N., Birkeland, A., Zhou, Q., Lu, G., Wang, H., Middendorf, L., Forouzanfar, T., Martin, B. A., Colevas, A., Rosenthal, E. L. 2019; 88: 58–65
  • In Vivo Optical Imaging of High-grade Glioma with Fluorescent EGFR Antibody World Molecular Imaging Congress (WMIC) Zhou, Q., Wilson, C., Vogel, H., Teraphongphom, N., Ertsey , R., Chu, P., Lu, G., van den Berg, N., van Keulen, S., Nishio , N., Grant, G. A., Rosenthal, E. 2018
  • Ultrasmall Paramagnetic Iron Oxide Nanoprobe Targeting Epidermal Growth Factor Receptor for In Vivo Magnetic Resonance Imaging of Hepatocellular Carcinoma. Bioconjugate chemistry Chen, Y., Zhou, Q., Li, X., Wang, F., Heist, K., Kuick, R., Owens, S. R., Wang, T. D. 2017; 28 (11): 2794-2803

    Abstract

    Hepatocellular carcinoma (HCC) is a common worldwide cancer that is rising rapidly in incidence. MRI is a powerful noninvasive imaging modality for HCC detection, but lack of specific contrast agents limits visualization of small tumors. EGFR is frequently overexpressed in HCC and is a promising target. Peptides have fast binding kinetics, short circulatory half-life, low imaging background, high vascular permeability, and enhanced tissue diffusion for deep tumor penetration. We demonstrate a peptide specific for EGFR labeled with an ultrasmall paramagnetic iron oxide (UPIO) nanoparticle with 3.5 nm dimensions to target HCC using T1-weighted MRI. We modified the hydrophobic core with oleic acid and capped with PEGylated phospholipids DSPE-PEG and DSPE-PEG-Mal. The EGFR peptide is attached via thioether-mediated conjugation of a GGGSC linker to the maleimide-terminated phospholipids. On in vivo MR images of HCC xenograft tumors, we observed peak nanoprobe uptake at 2 h post-injection followed by a rapid return to baseline by ∼24 h. We measured significantly greater MR signal in tumor with the targeted nanoprobe versus scrambled peptide, blocked peptide, and Gadoteridol. Segmented regions on MR images support rapid renal clearance. No significant difference in animal weight, necropsy, hematology, and chemistry was found between treatment and control groups at one month post-injection. Our nanoprobe based on an EGFR specific peptide labeled with UPIO designed for high stability and biocompatibility showed rapid tumor uptake and systemic clearance to demonstrate safety and promise for clinical translation to detect early HCC.

    View details for DOI 10.1021/acs.bioconjchem.7b00501

    View details for PubMedID 28972742

  • In vivo near-infrared imaging of ErbB2 expressing breast tumors with dual-axes confocal endomicroscopy using a targeted peptide. Scientific reports Gao, Z., Li, G., Li, X., Zhou, J., Duan, X., Chen, J., Joshi, B. P., Kuick, R., Khoury, B., Thomas, D. G., Fields, T., Sabel, M. S., Appelman, H. D., Zhou, Q., Li, H., Kozloff, K., Wang, T. D. 2017; 7 (1): 14404

    Abstract

    ErbB2 expression in early breast cancer can predict tumor aggressiveness and clinical outcomes in large patient populations. Accurate assessment with physical biopsy and conventional pathology can be limited by tumor heterogeneity. We aim to demonstrate real-time optical sectioning using a near-infrared labeled ErbB2 peptide that generates tumor-specific contrast in human xenograft breast tumors in vivo. We used IRDye800CW as the fluorophore, validated performance characteristics for specific peptide binding to cells in vitro, and investigated peak peptide uptake in tumors using photoacoustic tomography. We performed real-time optical imaging using a handheld dual-axes confocal fluorescence endomicroscope that collects light off-axis to reduce tissue scattering for greater imaging depths. Optical sections in either the vertical or horizontal plane were collected with sub-cellular resolution. Also, we found significantly greater peptide binding to pre-clinical xenograft breast cancer in vivo and to human specimens of invasive ductal carcinoma that express ErbB2 ex vivo. We used a scrambled peptide for control. Peptide biodistribution showed high tumor uptake by comparison with other organs to support safety. This novel integrated imaging strategy is promising for visualizing ErbB2 expression in breast tumors and serve as an adjunct during surgery to improve diagnostic accuracy, identify tumor margins, and stage early cancers.

    View details for DOI 10.1038/s41598-017-13735-z

    View details for PubMedID 29089571

    View details for PubMedCentralID PMC5663926

  • Visualizing epithelial expression in vertical and horizontal planes with dual axes confocal endomicroscope using compact distal scanner. IEEE transactions on medical imaging Li, G., Li, H., Duan, X., Zhou, Q., Zhou, J., Wang, T. 2017

    Abstract

    The epithelium is a thin layer of tissue that lines hollow organs, such as colon. Visualizing in vertical cross sections with sub-cellular resolution is essential to understanding early disease mechanisms that progress naturally in the plane perpendicular to the tissue surface. The dual axes confocal architecture collects optical sections in tissue by directing light at an angle incident to the surface using separate illumination and collection beams to reduce effects of scattering, enhance dynamic range, and increase imaging depth. This configuration allows for images to be collected in the vertical as well as horizontal planes. We designed a fast, compact monolithic scanner based on the principle of parametric resonance. The mirrors were fabricated using microelectromechanical systems (MEMS) technology and were coated with aluminum to maximize near-infrared reflectivity. We achieved large axial displacements [Formula: see text] and wide lateral deflections >20°. The MEMS chip has a 3.2×2.9 mm2 form factor that allows for efficient packaging in the distal end of an endomicroscope. Imaging can be performed in either the vertical or horizontal planes with [Formula: see text] depth or 1 ×1 mm2 area, respectively, at 5 frames/s. We systemically administered a Cy5.5-labeled peptide that is specific for EGFR, and collected near-infrared fluorescence images ex vivo from pre-malignant mouse colonic epithelium to reveal the spatial distribution of this molecular target. Here, we demonstrate a novel scanning mechanism in a dual axes confocal endomicroscope that collects optical sections of near-infrared fluorescence in either vertical or horizontal planes to visualize molecular expression in the epithelium.

    View details for DOI 10.1109/TMI.2017.2673022

    View details for PubMedID 28252391

  • Multimodal laser-based angioscopy for structural, chemical and biological imaging of atherosclerosis. Nature biomedical engineering Savastano, L. E., Zhou, Q., Smith, A., Vega, K., Murga-Zamalloa, C., Gordon, D., McHugh, J., Zhao, L., Wang, M., Pandey, A., Thompson, B. G., Xu, J., Zhang, J., Chen, Y. E., Seibel, E. J., Wang, T. D. 2017; 1

    Abstract

    The complex nature of atherosclerosis demands high-resolution approaches to identify subtle thrombogenic lesions and define the risk of plaque rupture. Here, we report the proof-of-concept use of a multimodal scanning fiber endoscope (SFE) consisting of a single optical fiber scanned by a piezoelectric drive that illuminates tissue with red, blue, and green laser beams, and digitally reconstructs images at 30 Hz with high resolution and large fields-of-view. By combining laser-induced reflectance and fluorescence emission of intrinsic fluorescent constituents in arterial tissues, the SFE allowed us to co-generate endoscopic videos with a label-free biochemical map to derive a morphological and spectral classifier capable of discriminating early, intermediate, advanced, and complicated atherosclerotic plaques. We demonstrate the capability of scanning fiber angioscopy for the molecular imaging of vulnerable atherosclerosis by targeting proteolytic activity with a fluorescent probe activated by matrix metalloproteinases. We also show that the SFE generates high-quality spectral images in vivo in an animal model with medium-sized arteries. Multimodal laser-based angioscopy could become a platform for the diagnosis, prognosis, and image-guided therapy of atherosclerosis.

    View details for DOI 10.1038/s41551-016-0023

    View details for PubMedID 28555172

    View details for PubMedCentralID PMC5446210

  • Targeted sections in either XY or XZ plane with dual-axes confocal endomicroscope Society of Photographic Instrumentation Engineers (SPIE) Photonics West Li, G., Li, H., Duan, X., Zhou, Q., Zhou, J., Oldham, K. R., Wang, T., 2017
  • Photoacoustic imaging of hepatocellular carcinoma targeting gold nanoshells Quan Zhou Zhou, Q., Chen, Y., Li, Z., Zhou, J., Duan, X., Wang, T., , 2017: 1007907
  • Novel Peptide Probe Targets GPC3 Overexpression in Vivo in Hepatocellular Carcinoma Quan Zhou Zhou, Q., Li, Z., Zhou, J., Duan, X., Kuick, R., Wang, T. 2017
  • Visualizing epithelial expression of EGFR in vivo with distal scanning side-viewing confocal endomicroscope SCIENTIFIC REPORTS Duan, X., Li, H., Zhou, J., Zhou, Q., Oldham, K. R., Wang, T. D. 2016; 6

    Abstract

    Confocal endomicroscopy is an emerging imaging technology that has recently been introduced into the clinic to instantaneously collect "optical biopsies" in vivo with histology-like quality. Here, we demonstrate a fast scanner located in the distal end of a side-viewing instrument using a compact lens assembly with numerical aperture of 0.5 to achieve a working distance of 100 μm and field-of-view of 300 × 400 μm2. The microelectromechanical systems (MEMS) mirror was designed based on the principle of parametric resonance and images at 5 frames per second. The instrument has a 4.2 mm outer diameter and 3 cm rigid length, and can pass through the biopsy channel of a medical endoscope. We achieved real time optical sections of NIR fluorescence with 0.87 μm lateral resolution, and were able to visualize in vivo binding of a Cy5.5-labeled peptide specific for EGFR to the cell surface of pre-cancerous colonocytes within the epithelium of dysplastic crypts in mouse colon. By performing targeted imaging with endomicroscopy, we can visualize molecular expression patterns in vivo that provide a biological basis for disease detection.

    View details for DOI 10.1038/srep37315

    View details for Web of Science ID 000388228500001

    View details for PubMedID 27874037

    View details for PubMedCentralID PMC5118792

  • In vivo fluorescence imaging of hepatocellular carcinoma xenograft using near-infrared labeled epidermal growth factor receptor (EGFR) peptide BIOMEDICAL OPTICS EXPRESS Li, Z., Zhou, Q., Zhou, J., Duan, X., Zhu, J., Wang, T. D. 2016; 7 (9): 3163-3169

    Abstract

    Minimally-invasive surgery of hepatocellular carcinoma (HCC) can be limited by poor tumor visualization with white light. We demonstrate systemic administration of a Cy5.5-labeled peptide specific for epidermal growth factor receptor (EGFR) to target HCC in vivo in a mouse xenograft model. We attached a compact imaging module to the proximal end of a medical laparoscope to collect near-infrared fluorescence and reflectance images concurrently at 15 frames/sec. We measured a mean target-to-background ratio of 2.99 ± 0.22 from 13 surgically exposed subcutaneous human HCC tumors in vivo in 5 mice. This integrated imaging methodology is promising to guide laparoscopic resection of HCC.

    View details for DOI 10.1364/BOE.7.003163

    View details for Web of Science ID 000385416500001

    View details for PubMedID 27699089

    View details for PubMedCentralID PMC5030001

  • Integrated monolithic 3D MEMS scanner for switchable real time vertical/horizontal cross-sectional imaging OPTICS EXPRESS Li, H., Duan, X., Qiu, Z., Zhou, Q., Kurabayashi, K., Oldham, K. R., Wang, T. D. 2016; 24 (3): 2145-2155

    Abstract

    We present an integrated monolithic, electrostatic 3D MEMS scanner with a compact chip size of 3.2 × 2.9 mm(2). Use of parametric excitation near resonance frequencies produced large optical deflection angles up to ± 27° and ± 28.5° in the X- and Y-axes and displacements up to 510 μm in the Z-axis with low drive voltages at atmospheric pressure. When packaged in a dual axes confocal endomicroscope, horizontal and vertical cross-sectional images can be collected seamlessly in tissue with a large field-of-view of >1 × 1 mm(2) and 1 × 0.41 mm(2), respectively, at 5 frames/sec.

    View details for DOI 10.1364/OE.24.002145

    View details for Web of Science ID 000371427100027

    View details for PubMedID 26906790

    View details for PubMedCentralID PMC5802237

  • Design and Synthesis of Near-Infrared Peptide for in Vivo Molecular Imaging of HER2 BIOCONJUGATE CHEMISTRY Joshi, B. P., Zhou, J., Pant, A., Duan, X., Zhou, Q., Kuick, R., Owens, S. R., Appelman, H., Wang, T. D. 2016; 27 (2): 481-494

    Abstract

    We report the development, characterization, and validation of a peptide specific for the extracellular domain of HER2. This probe chemistry was developed for molecular imaging by using a structural model to select an optimal combination of amino acids that maximize the likelihood for unique hydrophobic and hydrophilic interactions with HER2 domain 3. The sequence KSPNPRF was identified and conjugated with either FITC or Cy5.5 via a GGGSK linker using Fmoc-mediated solid-phase synthesis to demonstrate flexibility for this chemical structure to be labeled with different fluorophores. A scrambled sequence was developed for control by altering the conformationally rigid spacer and moving both hydrophobic and hydrophilic amino acids on the C-terminus. We validated peptide specificity for HER2 in knockdown and competition experiments using human colorectal cancer cells in vitro, and measured a binding affinity of kd = 21 nM and time constant of k = 0.14 min(-1) (7.14 min). We used this peptide with either topical or intravenous administration in a preclinical model of colorectal cancer to demonstrate specific uptake in spontaneous adenomas and to show feasibility for real time in vivo imaging with near-infrared fluorescence. We used this peptide in immunofluorescence studies of human proximal colon specimens to evaluate specificity for sessile serrated and sporadic adenomas. Improved visualization can be used endoscopically to guide tissue biopsy and detect premalignant lesions that would otherwise be missed. Our peptide design for specificity to HER2 is promising for clinical translation in molecular imaging methods for early cancer detection.

    View details for DOI 10.1021/acs.bioconjchem.5b00565

    View details for Web of Science ID 000370582600023

    View details for PubMedID 26709709

    View details for PubMedCentralID PMC5384256

  • . EGFR Targeting Photoacoustic Probe for Hepatocellular Carcinoma Imaging in Vivo Quan Zhou Zhou, Q., Li, Z., Zhou, J., Joshi, B., Li, G., Duan, X., Kuick, R., Owens, S., Wang, T., 2016: CTh2A-6
  • Vertical cross-sectional imaging of colonic dysplasia in vivo with multi-spectral dual axes confocal endomicroscopy. Gastroenterology Qiu, Z., Khondee, S., Duan, X., Li, H., Mandella, M. J., Joshi, B. P., Zhou, Q., Owens, S. R., Kurabayashi, K., Oldham, K. R., Wang, T. D. 2014; 146 (3): 615-617

    View details for DOI 10.1053/j.gastro.2014.01.016

    View details for PubMedID 24440675

  • Preparation and Characterization of a novel sludge conditioner by microbial flocculant (MBF) extracted from waste yeast with nano SiO2 particles Quan Zhou Zhou, Q., Zhou, C. 2011: 557-560