27

Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic
Page 2: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Welcome

http://mips.stanford.edu

he Molecular Imaging Program at Stanford (MIPS) was established as an interdisciplinary program to bring together scientists and physicians who share a common interest in developing and using state-of-the-art

imaging technology and developing molecular imaging assays for studying in-tact biological systems. A multimodality approach using imaging technologies such as positron emission tomography (PET), single photon emission comput-ed tomography (SPECT), digital autoradiography, magnetic resonance imag-ing (MRI), magnetic resonance spectroscopy (MRS), optical bioluminescence, optical fluorescence, photoacoustics, raman, and ultrasound are all technolo-gies under active development and investigation. The goals of the program are to fundamentally change how biological research is performed with cells in their intact environment in living subjects and to develop new ways to diagnose disease and monitor therapy in patients. Areas of active investigation include cancer research, microbiology/immunology, cardiovascular research, stem cell biology, quantitation and visualization, nanobiotechnology, early cancer detection, molecular probe development, developmental biology, and pharma-cology.

T

Sanjiv Sam Gambhir, MD, PhD Director, MIPSChair, Department of RadiologyVirginia and D.K. Ludwig Professor of Cancer ResearchProfessor by courtesy, BioengineeringProfessor by courtesy, Materials Science & EngineeringDirector, Canary Center at Stanford for Cancer Early DetectionMember, Bio-X Program

Gunilla Jacobson, PhD Deputy Director, MIPSMolecular Imaging Program at Stanford (MIPS)Department of RadiologyStanford School of Medicine

FACULTYDepartment

Raag Airan, MD, PhD Radiology (Neuroimaging and Neurointervention)

Sandip Biswal, MD Radiology

Francis Blankenberg, MD Radiology (Pediatric Radiology) & Pediatrics (courtesy)

Zhen Cheng, PhD Radiology

Frederick Chin, PhD Radiology

Heike E. Daldrup-Link, MD Radiology (Pediatric Radiology) & Pediatrics (courtesy)

Adam de la Zerda, PhD Structural Biology & Electrical Engineering (courtesy)

Dean Felsher, MD, PhD Medicine (Oncology) & Pathology

Sanjiv Sam Gambhir, MD, PhD Radiology, Bioengineering (courtesy), & Materials Science and Engineering (courtesy)

Edward Graves, PhD Radiation Oncology (Radiation Physics) & Radiology (courtesy)

Michelle James, PhD Radiology & Neurology and Neurological Sciences

Shivaani Kummar, MD Medicine (Oncology) & Radiology

Craig Levin, PhD Radiology, Physics (courtesy), Electrical Engineering (courtesy), & Bioengineering (courtesy)

Andreas Loening, MD, PhD Radiology (Body MRI)

Sanjay Malhotra, PhD Radiation Oncology (Radiation and Cancer Biology) & Radiology

Tarik Massoud, MD, PhD Radiology

Michael Moseley, PhD Radiology

Koen Nieman, MD, PhD Medicine (Cardiovascular Medicine) & Radiology (Cardiovascu-lar)

Ramasamy Paulmurugan, PhD Radiology

Guillem Pratx, PhD Radiation Oncology (Radiation Physics)

Jianghong Rao, PhD Radiology & Chemistry (courtesy)

Eben Rosenthal, MD Otolaryngology (Head and Neck Surgery) & Radiology

Brian Rutt, PhD Radiology

Daniel Spielman, PhD Radiology & Electrical Engineering (courtesy)

Avnesh Thakor, MD, PhD Radiology (Pediatric Radiology)

Juergen K. Willmann, MD Radiology (Body Imaging)

Joseph Wu, MD, PhD Medicine (Cardiovascular Medicine) & Radiology

Lei Xing, PhD Radiation Oncology (Radiation Physics) & Electrical Engineering (courtesy)

Page 3: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Research

http://mips.stanford.edu

What is Molecular Imaging?Molecular Imaging (MI) is a growing biomedical research discipline that enables the visualization, characterization, and quantification of biologic processes taking place at the cellular and subcellular levels within intact living sub-jects, including patients. MI images depict cellular and molecular pathways and mechanisms of disease present in the context of the living subject. Study of biologic processes in their own physiologically authentic environment is facilitated–MI transcends the requirements for and limitations of in vitro or ex vivo biopsy/cell culture laboratory techniques. It also encompasses ‘multiple’ image-capture techniques in combination with merging knowledge areas from the fields of cell/molecular biology, chemistry, pharmacology, medical physics, biomathematics, and bioinfor-matics.

Modern clinical scientist researchers use MI to study the processes of how molecular abnormalities, found in cells, build up to form the basis of disease. This type of study in turn facilitates other important clinical goals of 1) early detection of disease 2) optimizing therapies that aim for certain molecular targets 3) predicting and monitoring re-sponse to therapy and 4) monitoring for disease recurrence. Biotechnology companies also use MI to optimize the drug discovery and validation processes.

In stark contrast to classical imaging that details end-stage gross pathology/anatomy, MI reveals, within a living subject, the underlying biology occurring deep within cells anywhere in the body–this adds functional to anatomic information that is then available to clinical researchers and increases both our understanding about disease and our ability to intervene with treatment at an earlier time.

The Nuts and Bolts The Molecular Imaging Program at Stanford (MIPS) currently includes 28 different laboratories comprising 6 broad sections of MI technology and assay development in: Chemistry, Cell biology, Instrumentation, Pre-Clinical, Clinical, and Nano-technology. Within these MIPS laboratories, con-siderable research efforts are focused toward 5 key research areas that include:

1. synthesis and validation of molecular probes and nanoparticles for molecular imaging. 2. development of MI approaches/assays for in-terrogating cellular events in living subjects.3. development of MI instrumentation for living subjects.4. development of software tools for visualization and analysis of MI data.5. theranostics: merger of therapeutics and imag-ing strategies for improved patient management.

The science of the Molecular Imaging process, or ‘research chain’, that starts with a biological target of interest and spans to the actual imaging of a living subject is both complex and multi-faceted. This chain is fundamental to the field and critical for its success. Not all MI research is intended for clinical translation. In some cases, MI assay development progresses only to the computer modeling stage.

Technology DevelopmentMolecular Imaging originated in the field of nuclear medi-cine, and has developed to include an array of different strategies to produce imaging signals. Where nuclear medi-cine uses radiolabeled molecules (tracers) that produce sig-nals from radioactive decay only (PET or positron emission tomography, SPECT or single photon emission computed tomography), MI uses these and other molecules to image via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic particle imaging), or light (optical techniques of bioluminescence and fluorescence) as well as other emerging techniques e.g. Raman spectroscopy, optical coherence tomography (OCT), X-ray fluorescence tomography, and amide proton transfer imaging.

The adaptation/inclusion of ‘other’ MI technologies has oc-curred more recently through the development of different types of molecular probes. MI probes are classified at the widest level as: nonspecific or specific. Nuclear medicine plays a key role as specific probes, incorporating antibod-ies, ligands, or substrates, specifically interact with protein targets in particular cells or subcellular compartments–here the emphasis lies in imaging final products of gene expres-sion with radiolabeled substrates that interact with a pro-tein originating from a specific gene. These interactions are based on either receptor-radioligand binding, or enzyme mediated trapping of a radiolabeled substrate.

Due to the difficulties of ‘specific’ approaches (i.e., con-structing a different probe for each newfound target and then characterizing that probe in vivo), the development of nonspecific probes for ‘generalizable’ methods (i.e., those that can image gene product targets arising from the ex-pression of any gene of interest) has been inspired. This has further propelled the more recent development and vali-dation of MI reporter gene/reporter probe systems for use in living subjects.

History of Molecular Imaging and MIPS

As a field, Molecular Imaging dates back to the mid-1990’s for the start of its broad development. This start was enabled by a combination of factors–significant advances in molecu-lar/cell biology techniques, new methods of combinatorial drug design, high-throughput testing, and the notable emer-gence of novel imaging techniques and probes. Additional-ly, significant funding for the MI field became available from the NIH, and other agencies, in the late 1990’s.

MIPS was established as an interdisciplinary program in 2003 by the Dean of the School of Medicine (Dr. Philip Piz-zo) to bring together scientists and physicians who share a common interest in developing and using state-of-the-art imaging technology and developing MI assays for study-ing intact biological systems. The program, since its incep-tion, has been steadily directed by Dr. Sanjiv Sam Gamb-hir, Virginia & D.K. Ludwig Professor of Cancer Research and Chair of Radiology, and until 2016 co-directed by Dr. Christopher Contag, Professor of Pediatrics, and of Micro-biology and Immunology and by courtesy, of Radiology. To-day MIPS is directed by Dr. Gambhir together with Dr. Gu-nilla Jacobson as Deputy Director. Furthermore, Dr. Heike Daldrup-Link was appointed in 2017 as the Co-Director of the Cancer Imaging & Early Detection Program and in this capacity, serves as director of the Stanford Center for In-novation in In Vivo Imaging (SCI3).

MIPS has fostered a multimodality approach that uses imaging technologies such as positron emission tomogra-phy (PET), single photon emission computed tomography (SPECT), digital autoradiography, magnetic resonance imaging (MRI), magnetic resonance spectroscopy (MRS), optical bioluminescence, optical fluorescence, ultrasound, photoacoustic, Raman spectroscopy, and other emerging ones. All of these technologies are under active develop-ment and investigation. The founding and continuing goals of the MIPS program remain: i) to fundamentally change how biological research is performed with cells in their intact environment in living subjects, and ii) to develop new ways to diagnose diseases and monitor therapies in patients. Areas of active investigation span cancer research, micro-biology/immunology, neurology, cardiology, developmental biology and pharmacology. Funding for MIPS research ac-tivities comes from a mix of Federal (National Institutes of Health and Department of Energy), Foundation, and Uni-versity sources, as well as through numerous collaborations with industry.

Research chain

Page 4: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Research

http://mips.stanford.edu

microbubbles bound to a receptor called KDR found in the blood vessels of tumors that were malignant, but not in those that were benign.

Willmann JK, Bonomo L, Testa AC, Rinaldi P, Rindi G, Valluru KS, Petrone G, Martini M. Lutz AM, Gambhir SS. Ultrasound Molecular Imaging in Patients with Breast and Ovarian Lesions: First-in-Human Results. J Clin Oncol. 2017 Jul 1;35(19):2133-2140.

Detecting Cancers Through Tumor-Activatable Minicircles Leads to a Detectable Blood BiomarkerBlood-based cancer diagnosis is highly attractive, but current strategies suffer because they rely on the detection of en-dogenous molecules that often are secreted into the circulation by both malignant and nonmalignant cells. MIPS research-ers have shown that systemic administration of nonviral safe vectors they call “tumor-activatable minicircles” allows one to distinguish tumor-bearing from tumor-free subjects reliably and to assess tumor burden simply by measuring blood levels of such a reporter. This system represents an alternative paradigm for improved cancer detection and could enable more timely interventions to combat this devastating disease.

Ronald JA, Chuang HY, Dragulescu-Andrasi A, Hori SS, Gambhir SS. Detecting Cancers Through Tumor-Activatable Minicircles That Lead to a Detect-able Blood Biomarker. Proceedings of the National Academy of Sciences (USA). 2015; 112(10): 3068-73.

Visualizing Nerve Injury Leads to Pain Relief for PatientsThe ability to locate nerve injury and ensuing neuroinflammation will have tremendous clinical value for improving both the diagnosis and subsequent management of patients suffering from pain, weakness, and other neurologic phenomena associated with peripheral nerve injury. Current non-invasive techniques for assessing the clinical manifestations and morphological aspects of nerve injury often fail to provide accurate diagnoses due to limited specificity and/or sensitivity. Using a novel sigma-1 receptor (S1R) selective radioligand, [18F]FTC-146, and positron emission tomography-magnetic resonance imaging (PET/MRI) MIPS researchers can now accurately locate the site of nerve injury and impact how we diagnose, manage and treat patients with nerve injury. Clinical studies in humans are currently investigating the impact on several S1R-related diseases.

Shen B, Behera D, James ML, Reyes ST, Andrews L, Cipriano PW, Klukinov M, Lutz AB, Mavlyutov T, Rosenberg J, Ruoho AE, McCurdy CR, Gambhir SS, Yeomans DC, Biswal S, Chin FT. Visualizing Nerve Injury in a Neuropathic Pain Model with [18F]FTC-146 PET/MRI. Theranostics. 2017; 7(11): 2794-2805.

Shen B, Park JH, Hjørnevik T, Cipriano PW, Yoon D, Gulaka PK, Holly D, Behera D, Avery BA, Gambhir SS, McCurdy CR, Biswal S, Chin FT. Radiosyn-thesis and First-In-Human PET/MRI Evaluation with Clinical-Grade [18F]FTC-146. Mol Imaging Biol. 2017: 19 (5): 779-786.

Novel Technologies at MIPSTo fulfill the program goals, MIPS researchers have been working continuously to develop numerous key novel technologies, methods, and strategies for the advancement of molecular imaging. Some recent advances are de-scribed below.

A Novel Small Molecule Dye for NIR-II Fluorescence ImagingOptical imaging, such as fluorescence, offers the combination of both high spatial and temporal resolution along with the advantage of utilizing non-ionizing radiation. However, the reduced depth penetration has limited its use for clinical translation to endoscopy, ophthalmology, and dermatology. In an attempt to move more fluorescent probes to the clinic, MIPS researchers have been part of a team developing near infrared (NIR) emitting probes in the 1,000-1,700 nm range (the so-called second NIR window, or NIR-II) that are water-soluble, fast-excreting, and show much higher resolution than currently FDA approved NIR-I probes. Diminished tissue autofluorescence, reduced photon scattering, and low levels of photon absorption allow centimeter imaging depth at low resolution and micron-scale resolution of anatomic features (up to 3mm depth) that are otherwise unresolvable within the traditional NIR-I region (0.2mm depth).

Antaris AL, Chen H, Cheng K, Sun Y, Hong G, Qu C, Diao S, Deng Z, Hu X, Zhang B, Zhang X, Yaghi OK, Alamparambil ZR, Hong X, Cheng Z, Dai H. A small-molecule dye for NIR-II imaging. Nature Materials. 2016; 15(2): 235-42.

Hyperpolarized 13C MRS Provides In Vivo Real Time Window Into Metabolism

Hyperpolarized 13C magnetic resonance spectroscopy (MRS) is a functional imaging tool that allows us to study the reaction kinetics and enzyme activities in vivo via 13C-labeling of select metabolism. Dynamic nuclear polarization is used to hyperpolarize the 13C-labeled substrate before intravenous injection, and the metabolic activity can be fol-lowed to provide both spatial and chemical information. Until recently, hyperpolarized (13)C-pyruvate imaging stud-ies had focused solely on [1-(13)C]lactate production because of its strong signal. However, without a concomitant measure of pyruvate entry into the mitochondria, the lactate signal provides no information on the balance between the glycolytic and oxidative metabolic pathways. MIPS researchers have recently reported the reliable measure-ment of (13)C-bicarbonate production in both the healthy brain and a highly glycolytic experimental glioblastoma model, with the capacity to obtain signal in all brain tumors. This data suggests a potential application of this ratio as an early biomarker to assess therapeutic effectiveness. Additional new probes include [1-(13)C]glycerate for measuring glycolysis, and [1-(13)C]alanine for measurement of tissue redox state in the liver. [1-(13)C]pyruvate is currently being translated into new clinical trials for cancer research.

Park JM, Spielman DM, Josan S, Jang T, Merchant M, Hurd RE, Mayer D, Recht LD. Hyperpolarized (13)C-lactate to (13)C-bicarbonate ra-tio as a biomarker for monitoring the acute response of anti-vascular endothelial growth factor (anti-VEGF) treatment. NMR Biomed. 2016 May;29(5):650-9.

Ultrasound and Microbubbles Flag Malignant Cancer in HumansUltrasound complements mammography as an imaging modality for breast cancer detection, especially in patients with dense breast tissue, but its utility is limited by low diagnostic accuracy. MIPS researchers have developed an emerging molecular tool to address this limitation that involves contrast-enhanced ultrasound using microbubbles targeted to molecular signatures on tumor neovasculature, and demonstrated a way to diagnose cancer without resorting to surgery, raising the possibility of far fewer biopsies. In a recent first-in-human clinical trial, women with breast or ovarian tumors were injected intravenously with microbubbles capable of binding to and identifying cancer. The labeled microbubbles accumulated in the blood vessels of malignant tumors but not benign tumors. The labeled

Emerging Technologies Utilizing Magnetic Particle Imaging (MPI)Magnetic Particle Imaging (MPI) is an emerging molecular imaging technique that can non-invasively detect iron oxide nanoparticle tracers using time-varying magnetic fields. As the tracer is not normally found in the body, MPI images have exceptional contrast and high sensitivity. The technique has no signal attenuation with biological tissue and does not use ionizing radiation. MPI harnesses the flexibility of iron oxide nanoparticles as freely flowing through the vasculature, or to label cells as targeted probes. The technology has extensive applications for diagnostic imaging, such as: imaging can-cer, inflammation, tracking cellular therapeutics, and visualizing vascular perfusion. Several projects within MIPS are now utilizing this technology.

Song G, Rao J. Fluorescent iron oxide nanoparticles for magnetic particle imaging (MPI) of labeled cells and tumor xenografts of mice. 2017, WMIC, Philadelphia, Pennsylvania, Sept. 13-16. Paper in press.

Arami H, Teeman E, Troska A, Bradshay H, Saatchi K, Tomitaka A, Gambhir SS, Häfeli UO, Liggitt D, Krishnan KM. Tomographic magnetic particle imaging of cancer targeted nanoparticles. 2017, Nanoscale, DOI: 10.1039/C7NR05502A.

Page 5: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

Work in our lab focuses on the following areas: 1) the use of annexin V, a phos-phatidylserine binding protein, to reverse tumor immunosuppression in models of breast cancer; 2) the study of bacterio-phage imaging and therapy of pseudomo-

nas acute/chronic pulmonary infections in mice; 3) the use of Tc99m-HMPAO SPECT (a marker of intracellular glutathione) as an imaging biomarker of inherited and acquired mitochon-drial disease in the brain and other mitochondrial rich organs in children and adults; 4) application of bioinformatics / FTIR microspectroscopy in collaboration with Lawrence Berkeley National Laboratory Advance Light Source Division to study oxidative stress in live single cell fibroblasts and derived neu-rons from patients with inherited mitochondrial disease.

Francis Blankenberg, MDAssociate Professor, Radiology and Pediatrics

The Noninvasive Neurointerventions (ni2) Lab

is focused on developing novel molecular interventions for interrogating and treating the nervous system, primarily through focused ultrasound mediated targeted drug delivery. We are adapting the use of “phase-change”

nanotechnology to focally deliver neuromodulators to the brain to enable spatiotemporally-precise and receptor-specific noninvasive neuromodulation. In addition, we are implementing clini-cal protocols for targeted, safe, and reversible blood-brain barrier opening to increase chemotherapeutic delivery to the brain. Finally, we are exploring methods to use these technologies to focally modulate cerebral perfusion and the neural immuno-logical response.

Working in a multidisciplinary space that involves radiology, neurosurgery, neurology, and psychiatry, the ni2 lab is driven to

combine advances in drug delivery nanotechnology and focused ultrasound to enable noninvasive, spatially, and temporally precise drug delivery to the brain.

Chronic pain sufferers are, unfortunately, limited by poor diagnostic tests and thera-pies. Our lab is interested in the “imaging of pain” by using multimodality molecular imaging techniques to study molecular and cellular changes specific to nocicep-

tion and pain-ful inflammation as a means of improving objec-tive, image-guided diagnosis, and treatment of chronic pain disorders. We develop new molecular contrast agents for use in positron emission tomography (PET) and magnetic reso-nance imaging (MRI) and are currently conducting two clinical trials using the rela-tively new hybrid imaging technique of PET-MR. The overarching goals of our efforts is to develop an imaging approach that will pinpoint the exact cause of one’s pain, improve outcomes of pain sufferers, and to help develop new treatments for chronic pain.

Raag Airan, MD, PhDAssistant Professor, Radiology (Neuroimaging and Neurointervention)

Sandip Biswal, MDAssociate Professor, Radiology

The overall objective of this laboratory is to develop novel molecular imaging tech-niques and theranostic agents for early diagnosis and treatment of severe dis-ease, including cancer, neurological, and cardiovascular diseases. We have aimed

to identify novel cancer biomarkers with significant clinical relevance, explore new chemistry and platforms for imaging probe preparation, and develop new imaging strategies for clinical translation. To accomplish these goals, a multidisci-plinary team composed of members with expertise in organic chemistry, radiochemistry, biochemistry, bionanotechnology, molecular and cell biology, radiological science, medicine, and molecular imaging has been built to implement several research projects related to molecular imaging.

Zhen Cheng, PhDAssociate Professor, Radiology

Local drug delivery to the brain via focused ultrasound uncaging of nanoparticles

18F-FDG PET shows differential uptake of the radiotracer in the legs of human subjects with chronic leg pain (complex regional pain syndrome (CRPS), sciatica and osteo-arthritis (Sciatica/OA), peripheral nerve injury (PNI) and healthy volunteer (Normal)).

Imaging of cancer such as glioblastoma at NIR window II.

Noninvasive Neurointerventions Laboratory

Molecular Imaging of Nociception and Inflammation Laboratory

Nuclear Medicine Research Laboratory

Cancer Molecular Imaging Chemistry Laboratory

Page 6: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

High-Resolution Func-tional Imaging of Lym-phatic Drainage in Live Animals•This image of lym-phatic drainage and blood vasculature in a mouse ear was ob-tained using MOZART, a new high-resolution, high-sensitivity opti-cal imaging technique developed by the de la Zerda Lab. MOZART enables spectral differ-entiation of optimized gold nanorod contrast agents through the

implementation of custom image processing algorithms. In this experiment, MOZART was used to map discrete networks of draining lymph vessels con-taining different gold nanorod contrast agents (blue and green colors in the image) in virtual real-time. The detection algorithms also enable visualization of dense blood vessel networks in tissue (shown in red) and micro-anatomical structures. MOZART’s use of near infrared light for imaging enables micron-scale spatial resolution with millimeter-scale fields of view and depth of pen-etration into intact living tissues. Paired with high-sensitivity contrast agents, these features make MOZART an ideal technique for noninvasive molecular and functional imaging studies in living animals.

Our research team aims to provide pediatric patients with more efficient and accurate disease diagnoses than currently available. In the past, pediatric radiology essentially depicted human

anatomy. Modern medicine needs more advanced information. We combine innovations in nanoparticle development and medical imag-ing towards the development of novel imaging techniques which can detect specific cells in the body and monitor their function at a molecu-lar level. We developed novel imaging techniques for radiation-free cancer staging, imaging techniques for tracking of stem cell trans-plants in leukemia patients and “theranostic” (combined diagnostic and therapeutic) nanoparticles for image guided cancer therapy. A number of these molecular imaging technologies have been suc-cessfully translated from our basic science lab to clinical imaging applications, thereby creating direct value for our pediatric patients.

Heike Daldrup-Link, MDProfessor, Radiology & Pediatrics-Hematology/Oncology (courtesy)Director, Pediatric Molecular ImagingAssociate Chair, Diversity

This laboratory aims to build imaging in-strumentation and chemical tools that can visualize the complex behavior of biomol-ecules in living subjects. The expression patterns of many biomolecules (e.g., sig-naling factors and posttranslational modifi-

cations) changes in time, space and local environments. Un-derstanding these changes in the context of living tissues may give rise to new diagnostic and therapeutic approaches, and can further reveal new molecular mechanisms not otherwise visible in traditional biochemical studies. We have pioneered Photoacoustic molecular imaging and are actively develop-ing new optical imaging instrumentation to visualize these complex behaviors in cancer and ophthalmic disease animal models. Our research efforts span both basic science and clinically translatable work.

Adam de la Zerda, PhDAssistant Professor, Structural Biology

Combined inactivation of RAS and TWIST oncogenes results in de-creased lung tumor burden and FDG uptake.

Dean Felsher, MD, PhDProfessor, Medicine (Oncology) and PathologyDirector, Stanford Translational Research and Applied Medicine (TRAM) Program

My laboratory studies Oncogene Addiction.

We employ model systems whereby we can conditionally regulate oncogene expression in human or mouse cells in vitro or in mice. We

incorporate state-of-the-art methods of molecular imaging, and computational analysis to examine and model tumorigenesis.

We have a particular focus on examining when and how oncogene inactivation can be used to treat human cancer. Our work has uncovered the notion that tumors can be “oncogene ad-dicted”.

We have shown that oncogene addiction involves both tumor intrinsic as well as host (immune) dependent mechanisms.

We examine three questions:

1. How does oncogene activation cause cancer? 2. How and when does oncogene inactivation cause cancer to regress? 3. How can we predict when oncogene inactivation will cure cancer?

Pediatric Molecular Imaging Laboratory Cancer Biology Laboratory

Biophotonics Imaging Laboratory

Imaging Sigma-1 Receptors with Novel Radioligand [18F]FTC-146 First-in-Human study. Selected transverse brain PET at 30 min post injection

Frederick Chin, PhDAssistant Professor, Radiology

This lab specializes in synthetic chemistry and focuses on advancing radiopharmaceutical sci-ences for the expanding field of molecular imag-ing. We design and synthesize novel chemi-cal strategies that bind to various molecular targets related to specific neuropsychiatric disorders, pain, and cancer biology. In addition, new radiolabeling

techniques and methodologies are created in our lab for emerging radiopharmaceutical development as well as for the general radio-chemistry community. These radiochemistry approaches are coupled with innovative chemical engineering and in vivo models to further investigate new molecular imaging strategies. Successful imaging agents are also extended towards human clinical applications in-cluding disease detection and drug therapy.

Translational Radiopharmaceutical Sciences and Chemical Engineering Research (TRACER) for Molecular Imaging Laboratory

Page 7: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

Our clinical research in Medical Oncology is an integrated program that leverages the scientific and clinical expertise at Stanford. Phase I trials sit at the interface of labora-tory advances and later stage clinical de-

velopment; expedite development of new treatments while ensuring patient safety; and provide the basis to prioritize resource allocation and inform rational drug development strategies. The program conducts trials that provide proof of mechanism, proof of principle, and proof of concept early in the process of developing novel therapeutics. One of our re-search interests is to use imaging to evaluate drug pharmaco-kinetics and target modulation.

Michelle James, PhDAssistant Professor, Radiology & Neurology and Neurological Sciences

Shivaani Kummar, MDProfessor, Medicine and RadiologyDirector, Phase I Clinical Research Program

Edward Graves, PhDAssociate Professor, Radiation Oncology

This laboratory is focused on understanding tumor and normal tissue radiation response through the development and application of molecular imaging techniques. This goal is pursued through work spanning technique

development, basic research, and clinical translation. We are a multidisciplinary group with expertise in engineering, biol-ogy, chemistry, medicine, and computer science, and have developed a variety of methods of noninvasively detecting and quantifying molecular and physiologic aspects of radiation and tumor biology, including oxygen concentrations, hypoxia-regulated gene expression, metabolism, and cell migration. In addition, in order to evaluate the relevance of these molecular factors to clinical radiation therapy, we have developed a system for the delivery of clinically-similar image-guided conformal ra-diotherapy to small animals. This myriad of tools is being applied to elucidate the molecular, cellular, and clinical consequences of radiation exposure and cancer therapy.

Our lab is improving the diagnosis and treat-ment of brain diseases by developing translational molecular imaging agents for visualizing neu-roimmune interactions underlying conditions such as Alzheimer’s disease, multiple sclerosis, and

stroke. We are researching how the brain, its resident im-mune cells, and the peripheral immune system communicate at very early to late stages of disease. Our approach involves the discovery and characterization of clinically relevant immune cell biomarkers, followed by the design of imaging agents specifically targeting these biomarkers, and finally, the translation of promising probes to the clinic, enabling the precision targeting of immunomodu-latory therapeutics and real-time monitoring of treatment response in patients. We are passionate about our work, and excited about the impact these approaches will have in the lives of those suffering from debilitating brain diseases.

Radiation stimulates the migration of cancer cells both in vitro (left) and in vivo (right), as visualized using bioluminescence imaging.

Preclinical to clinical transition in early phase trials of novel anticancer agents.

Left panel: representative flow cytometry data validating a new myeloid-specific biomarker. Right panel: PET/CT imaging of this bio-marker using a novel tracer that our lab developed enabling detection and real-time tracking of the maladaptive immune response.

Sanjiv Sam Gambhir, MD, PhDChair, Department of RadiologyVirginia and D.K. Ludwig Professor of Cancer ResearchProfessor, by courtesy, Departments of Bioen-gineering and Materials Science & EngineeringDirector, Molecular Imaging Program at Stan-ford (MIPS)Director, Canary Center at Stanford for Cancer Early DetectionMember, Bio-X Program

The Multimodality Molecular Imaging Laboratory is developing imaging assays to monitor fundamental cellular/molecular events in living subjects, including patients. Technologies such as positron emission tomography (PET), optical (fluores-cence, bioluminescence, Raman), ultrasound, and photoacoustic imaging are all under active investigation. Imaging agents for multiple modalities including small molecules, engineered proteins, and nanoparticles are under development and being clinically translated. Our goals are to detect cancer early and to better manage cancer through the use of both in vitro diagnostics and molecular imaging. Strategies are being tested in small animal models and are also being clinically translated.

Multimodality Molecular Imaging Laboratory

Imaging Radiobiology Laboratory

Neuroimmune Imaging Research and Discovery Laboratory

Phase I Clinical Research Program

Spleen

LPS Veh K/O0

25

50

75

100

TREM

1+ (F

req

of P

aren

t %

)

Brain/Spinal Cord

Microglia (CD45mid CD11b+)

Myeloid biomarkerLymphoid (CD45+ CD11b-)

Myeloid (CD45+ CD11b+)

Other

Astrocytes (GFAP+)

Spleen

LPS Veh K/O0

25

50

75

100

TREM

1+ (F

req

of P

aren

t %

)

Biomarker Identification and Validation

PET Imaging of Maladaptive Inflammation in Multiple Sclerosis Mice

Spleen

EAE/MS Control

0

%ID

/g

28

Page 8: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

Our lab focuses on molecular and trans-lational imaging of the brain, especially in neuro-oncology. We develop experimen-tal and molecular imaging techniques for theranostic applications in brain cancer,

especially in glioblastoma (GBM), to interrogate cellular and molecular biological events, and to use in new anti-cancer therapeutic strategies. This includes in vivo imaging of gene expression using reporter assays, protein-protein interactions, and signal transduction, as well as cellular and nano-imaging. Other interests relate to animal modeling of gliomas, new glioma radiotracer development, studying the GBM p53 tran-scriptional network, imaging protein folding and misfolding in cancer, and developing novel nanoparticle-based drug and mi-croRNA formulations for ultra-targeted therapeutic strategies in endovascular neuro-oncology applications.

Tarik Massoud, MD, PhDProfessor, Radiology

Sanjay V. Malhotra, PhDAssociate Professor, Radiation Oncology & Radiology

Our laboratory focuses on the design and discovery of synthetic and natu-ral product-inspired small molecules, which can be used as probes to un-derstand biological phenomena, in-

cluding protein-protein interactions and modulation of signal transduction pathways. We employ the tools of synthetic and medicinal chemistry, molecular modeling, and chemical biology for translational research in drug discovery, development, imaging and radiation. Our cur-rent projects include design of new scaffolds/molecules as chemical tools to study various solid tumors, Alzheim-er’s disease, and markers for screening of hypoxic meta-bolically active cells. Administration of NRF2 activator TMC expands HSPCs by activating Notch1 signaling

in irradiated mice.

Small Molecule Design Laboratory

Laboratory of Experimental and Molecular Neuroimaging (LEMNI)

The lab focuses on research directed to-ward expanding the capability of MR and PET-MR as it relates to applications in body imaging. Clinical research aims in-clude the application of new or improved

MR sequences and reconstruction mechanisms to increase the speed, robustness, and diagnostic capability of body MR protocols, and combining PET molecular imaging agents with MRI to improve the diagnostic power of clinical imaging. Translational research aims include exploring new MRI con-trast mechanisms and contrast agents, such as for the strati-fication of cancer within the prostate and the identification of metastatic disease involvement of lymph nodes.

Andreas Loening, MD, PhDAssistant Professor, Radiology

Upper row: Conventional imaging (30 min scan time); Lower row: High-resolution post-contrast imaging (5 min scan time)Using a single high resolution post-contrast sequence allows faster imaging protocols and improved image assessment of rectal cancer. Example image demonstrates a patient with a large invasive rectal cancer and nodal metastases (orange arrow).

Body MR Translational Research Laboratory

The goal of the lab is to create novel instrumenta-tion and software algorithms for in vivo imaging of molecular signatures of disease in living sub-jects. These new cameras efficiently image emis-sions from molecular contrast agents to probe

disease biology in tissues residing deep within the body using mea-surements made from outside the body. The technology goals are to advance the sensitivity and spatial, spectral, and/or temporal reso-lutions, to create new camera geometries for special biomedical ap-plications, to understand the entire imaging process comprising the subject tissues, radiation transport, and imaging system, and to provide the best available image quality and quantitative accuracy. The ultimate goal is to introduce these new imaging tools into studies of molecular mechanisms and treatments of disease in living subjects.

Craig Levin, PhDProfessor, Radiology and by courtesy, Physics,Electrical Engineering, & Bioengineering

Schematic drawing of a “3-D” position sensitive scintillation detector concept used in the world’s first 1 millimeter resolution clinical PET system under con-struction in the molecular imaging instrumentation laboratory at Stanford.

Molecular Imaging Instrumentation Laboratory

Page 9: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

Guillem Pratx, PhDAssistant Professor, Radiation Oncology

The goal of the Radiation Biophysics labo-ratory is to create entirely new technolo-gies that address unanswered needs in the lab or the clinic. Our major focus is on ionizing radiation and how it can be used

for imaging and therapeutic applications. Combining a variety of physical approaches such as biomedical optics, radiation sensing, computing, nanotechnology, and microfabrication, our work spans a wide breadth of application, from answering biological questions at the single cell level to improving the accuracy of radiation treatments. For instance, the lab is part of the Medical Physics division, in Radiation Oncology.

Radiation Biophysics Laboratory

The objective of Dr. Nieman’s research is the development of accurate diagnostic techniques and more effective pathways to improve the management of patients with cardiovascular disease. Ongoing research

includes: (1) randomized controlled trials on tiered, compre-hensive cardiac CT protocols for stable chest pain, and CT angiography (CTA) for the triage of acute chest pain in the emergency room, (2) new functional cardiac CT applications such as stress myocardial perfusion imaging, and CTA-derived fractional flow reserve, (3) characterization of atherosclerotic plaque, (4) contrast media, and (5) 4D flow imaging with car-diac MRI.

The main focus of the CPIL is to develop in vivo imaging strategies to study cellu-lar signal transduction networks in cancer. Specifically, we study the signal transduc-tion networks involved in estrogen receptor (α and β)/hormone interactions, epigenetic

histone methylations, Nrf2-Keap1, Wnt-β-catenin, and NFkB-Nrf2 regulatory pathways and their roles in the pathogenesis and therapeutic responses of different cancers to various ther-apeutic interventions and drug resistance. Additionally, we de-velop microRNA mediated reprogramming approaches to en-hance cancer chemotherapy. With regard to breast cancer, we investigate the possible association of microRNAs with breast cancer development and tamoxifen resistance in particular. We also study signaling pathways to establish immunotherapy for cancer.

Koen Nieman, MD, PhDAssociate Professor, Medicine (Cardiovascular) & Radiology

Ramasamy Paulmurugan, PhDAssociate Professor, Radiology

Clinical Application of Advanced Cardiac Imaging

Cellular Pathway Imaging Laboratory

Michael Moseley, PhDProfessor, Radiology

Interests involve novel MR research for new thinking in the diagnosis of disease. MRI of tissue water is the best depiction of disease; mapping brain water diffusion has revolutionized our knowledge of the onset and evolution of ce-rebral stroke, making the MR scanner the Gold

Standard eyes and ears of choice for early and effective treatment of a variety of vascular diseases, trauma, cognition, and brain organization. MR tissue oxygenation allows us to ascertain oxygen utilization and metabolism. Up-to-date functional mapping can monitor neural networks while they work. Even minute physiologi-cal motions can be amplified with MR for a critical look at cellular density, pressures, and motions.

Cerebral blood volume plays a critical role in understanding brain dynam-ics, health, and response to stress. By visualizing even the smallest arteries and veins, surgeons can plan removal of malformations, avoid large feeding vessels to functional brain centers, or even map how a vascular stress such as carbon dioxide can affect normal brain from disease, for example. These maps can be made instantly and can be amplified to show subtle vessel dy-namics during the cardiac or respiratory cycle.

Research and Diagnosis of Disease from MR Imaging

Page 10: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

Medical imaging provides a wealth of infor-mation ranging from gross anatomy to bio-chemical processes. The Spielman labora-tory focuses on the development of novel methods for the non-invasive imaging of

metabolism and their translation to the clinic. Our current re-search efforts focus on using 1H magnetic resonance spec-troscopy (MRS) to measure neurotransmitter function and oxi-dative stress in the human brain and hyperpolarized 13C MRS and PET imaging to study glucose metabolism in both small animal models and clinical patients. Active collaborations in-clude studies of autism, schizophrenia, obsessive-compulsive disorder, cancer diagnosis and treatment monitoring, and met-abolic diseases including diabetes and non-alcoholic steato-hepatitis (NASH).

Daniel Spielman, PhDProfessor, Radiology & Electrical Engineering (courtesy)

The Spielman Laboratory for MRS and Multinuclear Imaging

This laboratory is interested in devel-oping and using in-vivo ultra-high field (e.g., 7T) Magnetic Resonance tech-niques to study human diseases. The increased sensitivity and enhanced contrast mechanisms at these high field

strengths should provide insight into unsolved problems, especially in neuroscience and cancer. Projects involve iron-loaded cell tracking, down to the single cell level, as well as the development and application of novel MR probes (contrast agents) for improved visualization and quantification of specific physiological as well as cellular and molecular processes.

MRI image of single metastatic cells in mouse brain. A) ME-labeled luc+231BR cells and B) Molday labeled luc+231BR cells. Mice were given an intracardiac injection of 1.5x105 cells prior to perfusion fixation. Brains were harvested and fixed overnight and then placed into a tube containing Fomblin prior to the MRI scan. A bSSFP MRI sequence was used to visualize single cells as signal voids (hypointensities).

Brian Rutt, PhDProfessor, Radiology

Ultra-High-Field Magnetic Resonance Imaging Research Laboratory

The Rosenthal lab focuses on develop-ment and clinical translation of novel im-aging probes and multimodal imaging strategies for improving cancer detection and treatment. Our recent research has

been mainly working towards first-in-human clinical trials us-ing near-infrared labeled antibodies (cetuximab, panitumumab) for surgical and pathological navigation during the surgery of head and neck cancer, brain cancer, and pancreatic cancer. We are also studying the role of optical imaging for quantification of antibody accumulation and distribution in the tissue and developing noninvasive imaging biomarkers to identify patients ame-nable to targeted therapy.

Eben Rosenthal, MDProfessor, Otolaryngology - Head and Neck Surgery, & Radiology

Translational Cancer Imaging Laboratory

The Rao lab is engaged in the quest for novel molecular imaging techniques to be ultimately de-ployed for patients at bedside, thus contributing to the detection and treatment of human diseases. Cost-effective, non-invasive, low dose molecular

probes are the tangible output and the reason to be part of the Rao lab, working both from the fundamental and applied standpoints. Among the latest research results accomplished, we must empha-size the development of a new approach, Target Enabled in Situ Li-gand Aggregation (TESLA), for detection of intracellular apoptosis in vivo, through a biocompatible condensation reaction, photoswitchable nanoparticles for background-removing photoacoustic imaging, and also the novel nanoparticle sensors for detection of reactive oxygen species induced by radiation therapy.

Jianghong Rao, PhDAssociate Professor, Radiology & Chemistry (courtesy)

Enzyme-directed synthesis and self-assembly of nanoparticles in living cells.

Cellular and Molecular Imaging Laboratory

Page 11: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Laboratories

http://mips.stanford.edu

This laboratory is focused on image instru-mentation, X-ray molecular imaging, image reconstruction, image processing, radia-tion therapy treatment planning, and image guided intervention. The group is develop-ing novel solutions to advance various clini-

cal imaging modalities such as CT, cone beam CT (CBCT), MRI, and PET, and investigating new strategies for molecular imaging and molecular image-guided therapeutics and treat-ment response assessment. We are also working on applica-tions of big data in radiation oncology and data-driven image analysis and treatment planning techniques.

Lei Xing, PhDProfessor, Radiation Oncology & Electrical En-gineering (courtesy)

The Xing Lab is studying Cerenkov Radiation Energy Transfer (CRET) of Gold Nanoclusters for molecular imaging. This figure illustrates the significant signal enhancement resulting from CRET over the conventional Cerenkov imaging technique (from Volotskova O, Sun C, Koh A, Pratx G, and Xing L, Efficient Radioisotope Energy Transfer (RET) by Gold Nanoclusters for Mo-lecular Imaging, Small 32,:4002-8, 2015).

Image Guided Intervention Laboratory

The Willmann lab develops and tests ultra-sound molecular imaging for identifying and monitoring diseases with the goal of us-ing this approach in the clinic for improved patient management. This novel imaging

modality uses intravascular contrast microbubbles which are modified to bind to regions of the diseased vasculature ex-pressing unique proteins. Using these microbubbles, we can detect small foci (<1 mm) of pancreatic and breast cancer and can monitor regions of diseased bowel undergoing active in-flammation. We have also successfully explored their use as a drug delivery vehicle for cancer therapy. Finally, our lab has performed the first-in-human clinical trial using these novel contrast agents in women with ovarian and breast cancer.

Juergen Willmann, MDProfessor, RadiologyVice Chair, Strategy, Outreach & Clinical TrialsChief, Body Imaging

(Top) On a contrast mode image obtained before contrast agent administration. (Bot-tom) Transverse contrast mode image obtained at 11 minutes after intravenous ad-ministration of KDR- targeted contrast microbubbles (MBKDR) show strong and per-sistent targeted ultrasound image signal in breast cancer and low background signal.

Joseph Wu, MD, PhDDirector of Stanford Cardiovascular InstituteSimon H. Stertzer, MD Professor of Medicine (Cardiology) & Radiology

The Wu lab studies the biological mecha-nisms of adult stem cells, embryonic stem cells, and induced pluripotent stem cells. We use a combination of next generation sequencing, tissue engineering, physi-

ological testing, and molecular imaging technologies to better understand stem cell biology in vitro and in vivo. For adult stem cells, we are interested in monitoring stem cell survival, proliferation, and differentiation. For embryonic stem cells, we are currently studying their tumorigenicity, immunogenicity, and differentiation. For induced pluripotent stem cells, we are interested in cardiovascular disease modeling, drug screening, and cell therapy. We also develop novel vectors and therapeutic genes for cardiovascular gene therapy applications.

Imaging of human induced pluripotent stem cell-derived cardiomyocytes.

Cardiovascular Stem Cell Laboratory

Translational Molecular Imaging Laboratory

Our laboratory’s research primarily focuses on the pancreas. We conduct research related to diabe-tes by investigating beta cell regeneration using mesenchymal stem cells with the use of pulsed focused ultrasound for mesenchymal stem cell

homing, islet cell transplantation, and the construction of novel “ac-tive” bioscaffolds for islet transplantation. We also study pancreatic cancer by developing novel intra-arterial delivery techniques to the pancreas and the synthesis of theranostic nanoparticle platforms.

Avnesh S. Thakor, MD, PhDAssistant Professor, Radiology

A novel strategy which combines bioscaffolding and stem cell coating for islet transplantation.

Interventional Regenerative Medicine and Imaging Laboratory

Page 12: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Facilities

http://mips.stanford.edu

http://mips.stanford.edu/aboutus/facilities/sci3.html

The mission of the Stanford Center for Innovation in In vivo Imaging (SCI3) is to provide access to state-of-the-art pre-clinical imaging instruments to permit easy translation of re-search from in vitro procedures (tissue culture, etc.) to small animal models. This permits the application and advance-ment of technologies for biological assessment and imaging in living murine models. Researchers are trained in use of each of the many imaging modalities available, and several facilities are located on campus to enable ease of access for scientists and their research animals.

The facility provides access to many imaging modalities, including instruments routinely found in hospitals, but opti-mized for small animal work (such as ultrasound, MRI, Mi-croCT, PET and SPECT), instruments developed specifical-ly for small animal work (such as optical imaging systems capable of bioluminescent and fluorescent imaging), as well as new technologies where equipment has just been de-veloped and installed (such as photoacoustic imaging and magnetic particle imaging). All instruments are designed to image living subjects, so repeated and longitudinal stud-ies can be performed, which provide better data as well as reducing the number of animals required for such studies. The flexibility and rapid analyses of such animal models greatly accelerate the development of molecular imaging strategies, as well as new therapeutic strategies for a vari-ety of diseases.

The Clark Center houses the primary SCI3 facility, with sep-arate imaging centers located in the Lorry I. Lokey Stem Cell Research Building (SIM1), Comparative Medicine Pa-vilion, Shriram Center for Bioengineering & Chemical Engi-neering, and the Porter Drive facility off campus.

The SCI3 is a Stanford School of Medicine service center, and is supported by the Stanford Cancer Institute, as well as user fees levied on each instrument. It is operated by the Departments of Pediatrics and Radiology.

Stanford Center for Innovation in In vivo Imaging (SCI3)

Tim Doyle, PhDScientific Director, Clark Center Facility

Heike Daldrup-Link, MDDirector, Pediatric Molecular Imaging

Laura Jean Pisani, PhDDirector, Pre-clinical MRI

Frezghi Habte, PhDDirector, Preclinical Imaging Facility at Porter

Page 13: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Facilities

http://mips.stanford.edu

http://med.stanford.edu/mips/aboutus/facilities/radiochem istry.html

The Cyclotron and Radiochemistry Facility (CRF) devel-ops and delivers radioactively-labeled imaging probes, also called radiotracers, for use in early detection, therapeutic monitoring, and theranostic treatment of disease. These ra-diotracers are used to support clinical imaging scans (such as PET and SPECT) as well as research studies at the Stanford Hospital, the Lucile Packard Children’s Hospital, and the Stanford Center for Innovations in In Vivo Imaging (SCI3). Radiotracers are injected into living subjects during a PET or SPECT scan to noninvasively visualize internal biological targets of interest; many of these radiotracers are applied in the areas of oncology, cardiology, and neurologi-cal diseases.

The CRF is the main radiochemistry facility at Stanford with the primary mission of providing expertise in the de-sign, synthesis, and production of current and new imaging probes. Leadership of the facility is provided by Dr. Fred-erick T. Chin (Director, CRF since 2005) and Dr. Bin Shen (Manager, CRF since 2016). In total, nearly 30 radiochemis-try personnel (including students, staff, and faculty) operate this facility daily and support its mission.

Cyclotron: The heart of the CRF is a 16.5 MeV GE PETtrace 880 cyclotron which is used for the production of radioisotopes for both clinical and research use. The cyclo-tron runs on demand to support delivery of 18F-, 11C-, 13N-, and 15O-isotopes as needed for each day’s radiochemis-try schedule. In addition, the CRF can provide longer-lived isotopes (e.g., 68Ga, 64Cu, and 89Zr). Timely delivery of ra-diotracers is the essential final step in the CRF operation, especially since several routinely-used clinical radiotracers have half-lives of approximately 110 minutes or less; us-ing radiotracers beyond their designed timeframe (due to radioactive decay and required molar activity) renders them ineffective for clinical or research use.

Cyclotron and Radiochemistry Facility

Clinical radiotracer production: Adjacent to the cyclotron is the GMP production facility, equipped to synthesize rou-tine radiotracers while abiding by the current regulatory poli-cies. Since 2006, the CRF continues to provide 18F-FDG to Stanford Hospitals and Clinics for patient standard-of-care (approximately 5,500 doses/year) and will begin serving other newly-acquired satellite Stanford Hospitals in the Bay Area. To date, more than 30 tracers (with many others cur-rently pending under FDA/RDRC review) can be ordered from the CRF for clinical use, clinical research, or clinical tri-als. The number of available tracers has grown significantly over the past 12 years and is a statement of the dedication of the CRF to meet the needs of patients and its commit-ment to innovation in developing new imaging methodolo-gies.

Pre-clinical research and translation: In addition to radio-tracer production for clinical use, the CRF includes space for hot labs, fully equipped for research and development of new radiotracers. This key facet of the CRF supports the vision of the Molecular Imaging Program at Stanford (MIPS) which was established in 2003 as an interdisciplinary ini-tiative at Stanford Medicine. The goal of these efforts is to advance molecular imaging of living subjects by providing state-of-the-art molecular imaging strategies to improve our understanding of the in vivo biological events during dis-ease progression and to focus on clinical translation for im-proved patient care.

CRF Personnel (From left to right): Zheng Miao, Jessa Castillo, George Montoya, Jun Hyung Park, Frederick Chin, Shawn Scatliffe, Murugesan Subbaryan, Carmen Azevedo, and Bin Shen.

Page 14: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Facilities

http://mips.stanford.edu

http://med.stanford.edu/lucasmri.html

The Richard M. Lucas Center for Imaging is an integral part of the department of Radiology and the divisions of MIPS and Radiological Sciences Laboratory (RSL). Its primary mission is to advance imaging in healthcare at Stanford through technology creation and development, translational research and education. It houses resources devoted to research in magnetic resonance imaging (MRI), spectroscopy (MRS), positron emission tomography (PET) and X-ray/CT imaging. Re-searchers at the Lucas Center have pioneered MRI/MRS/X-ray/CT technology while developing new techniques that benefit patients with stroke, cancer, heart disease and brain disorders. The Center supports collaborative and original research us-ing human subjects and intact animal models.

Our facilities include two 3.0T whole-body MR systems, a 3.0T whole-body PET/MR system and a 7.0T whole-body MR system, complete with patient/animal preparation facilities and image processing/ readout workstations. The Axiom/Zeego Lab houses a C-arm X-ray CT scanner that offers fluoroscopic scanning, and many other applications. A machine shop/ workshop offers addition capability for miscellaneous hardware development including MRI coils and phantoms.

3T MR systems (GE Healthcare: Signa-Premier, MR750): The Signa-Premier system is a wide bore (70cm) MR sys-tem with very high performance gradients (strength: 50 mT/m and slew rate: 200 mT/m/ms) and is the second such installation world-wide. The 3T MR systems include multi-nuclear capability, high performance gradients and a wide range of RF coils to enable a variety of cutting edge re-search. The scan and control rooms support many applica-tions with multiple workstations and stimulus equipment for fMRI studies.

3T PET/MR system (GE Healthcare: Signa PET/MR): The simultaneous PET/MRI scanner combines the high spatial resolution and fine anatomical soft tissue detail of MRI with the high molecular specificity of PET imaging, in real-time, providing true functional imaging capabilities. Moreover, the Time-of-Flight (TOF) PET provides improved Signal-to-Noise Ratio (SNR) and Carrier-to-Noise Ratio (CNR), thereby reducing the radiotracer dosage required. Some of the novel PET/MR research applications pursued include adult and pediatric oncology, neurodevelopmental disor-ders, neurodegenerative disorders and pain imaging.

7T MR system (GE Healthcare: MR950): The system includes a 32-channel RF head coil (Nova Medical) and a selection of multinuclear coils for neuroimaging and musculoskeletal imaging. Some of the research projects involve the development of basic technology of MRI (e.g., insertable gradient coils and RF components), basic science applications of MR (e.g., in vivo detection of rare cell populations using MRI) and clinical applications (e.g., multiple sclerosis). The increased sensitivity and enhanced contrast mechanisms at these high magnetic field strengths provide novel insights into unsolved problems, especially in neuroscience and cancer.

The Center also houses a mock scanner used to introduce the MR examination environment to special patient populations including children, the elderly, those prone to claustrophobia, and other conditions. It has complete facilities for preparation of animal models, wet labs, data analysis laboratories, and an electronics/RF coil workshop. Much of the equipment is oper-ated as a University Service Center, available to Stanford and non-Stanford researchers by arrangement with the Center administrator.

The Richard M. Lucas Center for Imaging

Page 15: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Research Grants

http://mips.stanford.edu

(CCNE-TD) - NCI U54Sanjiv Sam Gambhir, MD, PhD, Principal InvestigatorShan X. Wang, PhD, Co-Principal InvestigatorDemir Akin, PhD, Deputy Director http://med.stanford.edu/ccne/ccne-td/

The Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), which forms the third cycle CCNE Program at Stanford University, is a consortium that has three highly synchronized Projects and three Cores. Since its initial funding in May 2006, the CCNE program has matured substantially into a strong multidisciplinary program with expertise and infrastructure to support the growing field of cancer nanomedicine.

The Center is composed of a highly interdisciplinary team of scientists whose expertise areas are highly synergistic and have a long collaboration history that extends to the first cycle of the NCI’s CCNE Program. Defining it broadly, the CCNE-TD is developing and clinically translating cancer diagnostics and imag-ing technologies. More specifically, the Center has two scientific thematic focus areas: i) predicting and monitoring cancer therapy response in lung cancer and ii) merging of nano-based in vitro and in vivo diagnostics strategies as well as nano-based imaging for earlier cancer detection and prognostication for prostate cancer. The Projects (P) and Cores (C) are as follows: P1 (Lead: Jianghong Rao, PhD) focuses on the development of novel cancer triggered self-assembling and disas-sembling nanoparticles for photoacoustic and PET-MRI visualization of tumors, P2 (Lead: Shan X. Wang, PhD) focuses on the use of magneto-nanotechnology for blood proteomics, single cell sorting and comprehensive analyses, P3 (Lead: San-jiv Sam Gambhir, MD, PhD) focuses on molecular imaging of prostate cancer with photoacoustics smart nanoparticles that will be made by Project 1, and monitoring response to anti-lung cancer therapy using imaging and magneto-nanosensors. C1 (Lead: Demir Akin, DVM, PhD) is the Administration Core and facilitates prog-ress towards our milestones, C2 (Lead: Robert Sinclair, PhD) provides resources for nanocharacterization and nanofabrication, C3 (Lead: Alice Fan, MD) facilitates clinical translation by linking our nanotechnologies to existing patient samples and ongoing as well as new clinical trials.

Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD)

Overall vision of the Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD)

The CCNE-TD investigators are utilizing nanotechnology to measure changes in cancer patterns via 1) imaging though cancer-triggered-self-assembling as well as dis-assembling nanoparticles (P1 and P3) and 2) in the serum using mag-neto-nano sensors (P2). Our nanotechnologies are used to interrogate single cells for DNA, RNA, proteins, cellular mi-cro/nano vesicles, to evaluate biomarker potentials of those components (P2, P3). We are again utilizing nanotechnol-ogy (e.g., self-assembling nanoparticles and nanobubbles) to image cell associated proteins in small animal models, as well as humans, and clinically translate them for human prostate cancer imaging with ultrasound/photoacoustics (P3). In all of these cases, changes at the molecular level are being measured within the cell, on the cell membrane, and in the extracellular matrix. Measuring these changes is critical to the problem of earlier cancer detection and moni-toring response to therapies with both the ex vivo diagnostic nanosensor technologies and the in vivo imaging technolo-

gies. The Center has two major technological arms: i) in vitro genomic/proteomic/cellomic nanosensors, and ii) in vivo molecular imaging with primarily gold as well as nano-bubble-based nanoparticles, and magnetic resonance im-aging (MRI) with novel self-assembling and dis-assembling nanoparticles. The latter arm is directly focused on molecu-lar imaging with specific cellular protein targets. These tar-gets are the basis whereby specific molecular imaging sig-nal is provided. It is the goal of the CCNE-TD to help identify these targets for specific cancers (lung and prostate) and their biochemical pathways and to utilize these targets as ways to home in on cancer cells. In addition, a new class of nanoparticles that can self-assemble intracellularly, from the cell permeable precursors, in a cancer specific manner, in conjunction with advanced magnetic resonance imaging (MRI) expertise, is expected to directly impact the devel-opment of medical imaging modalities for eventual clinical translation.

Page 16: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

http://mips.stanford.edu

Cardiovascular Imaging Postdoctoral Fellowship Training at Stanford (CVIS)

(CVIS) - NIH NIBIB T32 Joseph C. Wu, MD, PhD, Program Director John M. Pauly, PhD, Co-Director Koen Nieman, MD, PhD, Co-Director http://med.stanford.edu/cvi/education/cvis-t32.html

The Multidisciplinary Training Program in Cardiovascular Imaging at Stanford (CVIS) is funded by the National Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health. The program is designed to train the next generation of CV imaging inves-

tigators by exposing them to three complementary areas–clinical, engineering, and molecular imaging. With the impact of cardio-vascular disease on U.S. and world health and the rapid advances in imaging technologies and cardiovascular biology, it is critical that fellows be provided a broad, multidisciplinary, and collaborative training program to foster their ability to translate CV imaging research into clinical application.

Cancer-Translational Nanotechnology Training (Cancer-TNT) Program

(Cancer-TNT) - NCIJianghong Rao, PhD, Program DirectorDean Felsher, MD, PhD, Co-Director http://med.stanford.edu/ctnt.html

The Cancer-Translational Nanotechnology Train-ing (Cancer-TNT) Program is led by Drs. Jiang-hong Rao and Dean Felsher and is funded by the

National Cancer Institute (NCI). The vision of this three-year postdoctoral fellowship program is to train a new generation of scien-tists with the skills and educational backgrounds from the areas of engineering, chemistry, materials science, cancer biology and medicine. This program will provide the opportunity for talented scientists to learn the intricacies of merging nanotechnology with the biological and medical sciences, specifically for use in cancer, in addition to becoming leaders in the rapidly growing field of cancer nanotechnology.

Advanced Residency Training at Stanford (ARTS) Program

Sanjiv Sam Gambhir, MD, PhD, Program Directorhttp://med.stanford.edu/arts.html

In addition to the NIH-funded training programs, Stanford is also home to the Advanced Residency Training at Stanford (ARTS) Program that offers residents and clinical fellows the opportunity to combine their clinical training with advanced research training to complete a PhD degree during

or upon completion of residency or clinical fellowship. The program begins with one or more years of postgraduate clinical training, followed by research training in one of twenty graduate programs from the Schools of Medicine, Engineering, or Humanities and Sciences. Through the ARTS Program that provides individuals with the tools needed to move freely between the laboratory and the clinic, Stanford demonstrates its commitment to the emerging disciplines of translational medicine and precision medical care.

The MINDED Programme, a Marie Skłodowska-Curie (MSC) COFUND Action

European Union’s Horizon 2020 Paolo Decuzzi, PhD, IIT Program DirectorBrian Rutt, PhD, MIPS, Stanford, Co-Directorhttp://minded-cofund.eu/

The Marie Skłodowska-Curie programme MINDED offers 24 prestigious 4-year fellowships for experienced researchers with the objective of advancing the diagnosis, imaging and treatment of neurodevelop-mental disorders. Selected fellows will be trained for business development and clinical translation of new biomedical technologies, and will originate a new class of scientists and entrepreneurial scientists. MINDED is led by the Istituto Italiano di Tecnologia (Italian Institute of Technology – IIT) in Genoa (Italy) and involves 16 partners in Europe, USA and Israel, comprising 11 research centers and universities and 6 non-academic institutions.

MIPS and Stanford University is the only USA based partner in MINDED, and will host several MINDED trainees for 2-4 years. Besides being part of the MIPS program these trainees will interact and collaborate with other MINDED partners and visit IIT annually for joint research meetings.

Stanford Cancer Imaging Training (SCIT) Program

(SCIT) - NCISandy Napel, PhD, Co-DirectorBruce Daniel, MD, Co-Director http://scitprogram.stanford.edu/

The Stanford Cancer Imaging Training (SCIT) Program, funded by the National Cancer Institute, aims to train the next generation of researchers in the develop-

ment and clinical application of advanced techniques for cancer imaging. Our coursework, rich mentored training opportunities, and out-standing resources, provide an active, vibrant program that attracts students nationwide. Graduates from our program are highly sought after, filling faculty and industry research positions internationally.

The SCIT program is surrounded by and draws from numerous departments, programs, and resources with core relevant competencies to imaging science. Trainees will have access to our exceptional imaging facilities, including the Richard M. Lucas Center for Imaging in the Department of Radiology, as well as additional research space in more than 10 other buildings throughout the School of Medicine. The SCIT Program also facilitates unique research collaborations with the Bio-X Program, the Center for Biomedical Imaging at Stanford (CBIS), the Molecular Imaging Program at Stanford (MIPS), the Radiological Sciences Laboratory (RSL), and the Integrative Biomedical Imaging Informatics at Stanford (IBIIS).

Stanford Molecular Imaging Scholars (SMIS) Program

(SMIS) - NIH R25TCraig S. Levin, PhD, Program Directorhttp://smisprogram.stanford.edu/

The Stanford Molecular Imaging Scholars (SMIS) program is a diverse train-ing program bringing together more than thirteen Departments, predominant-

ly from the Stanford Schools of Medicine and Engineering, in order to train the next generation of interdisciplinary leaders in molecular imaging. Oncologic molecular imaging is a rapidly growing area within molecular imaging which combines the disciplines of chemistry, cell/molecular biology, molecular pharmacology, physics, bioengineering, imaging sciences, and clinical medicine to advance cancer research, diagnosis and management.

The goals of SMIS are to train postdoctoral fellows through a diverse group of over 40 basic science and clinical faculty mentors repre-senting 8 program areas, incorporating formal courses in molecular imaging, molecular pharmacology, cancer biology, cancer immunol-ogy, virology, and gene therapy, with a clinical component including hematology/oncology rounds.

Training Grants

Page 17: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

http://mips.stanford.edu

EducationMolecular Imaging Related Courseshttp://med.stanford.edu/mips/education.html

Journal ClubsMIPS Journal Club

Every other Thursday, 12:00 pm–1:00 pm Clark Center, Room S-363 http://med.stanford.edu/mips/events/journal_club

Nuclear Medicine & Molecular Imaging Clinical Journal Club

First Wednesday of every month, 11:00 am–12:00 pm Nuclear Medicine Library, Room H2211

Seminar SeriesCME Radiology Grand Rounds

Global Learning Objectives:

1. Critically analyze research, guidelines and appropriate use criteria to develop best-practice diagnosis and treat-ment strategies. 2. Evaluate latest innovations in imaging to assess safety and effectiveness.

Li Ka Shing Center, LK 120/130 /Clark Center Auditorium 1st Tuesdays, 7:30 am – 8:30 am (except June, July, & August) 3rd Thursdays, 5:30pm–6:30pm (except June, July, & Au-gust)

http://radiology.stanford.edu/education/grandrounds.html

IMAGinING the Future

Quarterly seminars by world-renowned scientists aimed at catalyzing interdisciplinary discussions in all areas of medi-cine and disease.

Wednesdays, 1:00 pm–2:00 pm, Berg Hall, Li Ka Shing Center http://med.stanford.edu/mips/imagining-the-future.html

Early Detection Seminar Series

Bimonthly seminars highlighting new research in Early Can-cer Detection. Seminar & Discussion: 4:30pm–5:30pm Reception: 5:30pm–6:00pm http://canarycenter.stanford.edu/seminars/early-detection.html

Integrative Biomedical Imaging Informatics at Stanford (IBIIS) Seminar Series

Monthly seminar series, 3:30 pm–4:30 pm http://ibiis.stanford.edu/events/seminars.html

PHIND Seminars

Monthly seminar series Clark Auditorium http://med.stanford.edu/phind/events.html

SCIT Program Seminars

Quarterly colloquium presentations by trainees Lucas Expansion, Glazer Learning Center http://med.stanford.edu/scitprogram/seminar.html

Medical Imaging Seminar

Occasional Wednesdays, 10:00 am–11:00 am Li Ka Shing Center http://cbis.stanford.edu/events/MIseminar.html

BioEngineering 221 / Radiology 221Physics and Engineering of Radionuclide Imaging

Physics, instrumentation, and algorithms for positron emission tomography (PET) and single photon emission comput-ed tomography (SPECT). Topics include basic physics of photon emission and detection, electronics, system design, strategies for tomographic image reconstruction, data correction algorithms, methods of image quantification, image quality assessment, and current developments in the field.

BioEngineering 222 / Radiology 222Instrumentation and Applications for Multimodality Molecular Imaging of Living Subjects

Focuses on instruments, algorithms and other technologies for imaging of cellular and molecular processes in living subjects. Introduces preclinical and clinical molecular imaging modalities, including strategies for molecular imaging using PET, SPECT, MRI, Ultrasound, Optics, and Photoacoustics. Covers basics of instrumentation physics, the origin and properties of the signal generation, and image data quantification.

BioEngineering 224 / Radiology 224Probes and Applications for Multimodality Molecular Imaging of Living Subjects

Focuses on molecular contrast agents (a.k.a. “probes”) that interrogate and target specific cellular and molecular disease mechanisms. Covers the ideal characteristics of molecular probes and how to optimize their design for use as effective imaging reagents that enables readout of specific steps in biological pathways and reveal the nature of disease through noninvasive imaging assays.

BioEngineering 229Advanced Research Topics in Multimodality Molecular Imaging of Living Subjects

Covers advanced topics and controversies in molecular imaging in the understanding of biology and disease. Lectures will include discussion on instrumentation, probes and bioassays. Topics will address unmet needs for visualization and quantification of molecular pathways in biology as well as for diagnosis and disease management. Areas of unmet clinical needs include those in oncology, neurology, cardiovascular medicine and musculoskeletal diseases.

Radionuclide Imaging Basic Science Lectures

Fridays, 12:00 pm–1:00 pm Nuclear Medicine Library, H2211

Nuclear Medicine & Molecular Imaging Clinical Lecture Series

Mondays, 12:00 pm–1:00 pm Nuclear Medicine Library, H2211

Nuclear Medicine & Molecular Imaging Clinical Case Conference

Wednesdays, 11:00 am–12:00 pm Nuclear Medicine Reading Room

Page 18: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

New Initiatives

http://mips.stanford.edu

The New Lucile Packard Children’s HospitalLucile Packard Children’s Hospital is expanding its walls in order to meet growing community needs for specialized pedi-atric and obstetric care. In order to serve this growing patient base, an expanded facility adjacent to the current Packard Children’s Hospital was built and opened in November 2017.

As part of Packard Children’s mission to provide family-cen-tered care, the completed Packard Children’s Hospital Ex-pansion features additional single-patient rooms and more space for families to be with their child during treatment and recovery. The expansion provides patients and doctors with the most modern clinical advancements and technology while also addressing the specialized needs of pediatric and obstet-ric patients and their families.

The New Stanford HospitalStanford Hospital & Clinics is rebuilding its 1950s-era hospital facilities to accommodate new medical technology, increase capacity needs and meet seismic-safety requirements. The new facilities feature individual patient rooms, an enlarged Level-1 trauma center and Emergency Department and new surgical, diagnostic and treatment rooms. The rebuild of Stan-ford Hospital is the next step in the hospital’s successful 50 year history. The new facilities are leading the way for con-tinued success in providing advanced patient care and treat-

ment to the surrounding communities. The hospital will remain open and fully operational throughout construction of the new facilities.

Hyperpolarized 13C Magnetic Resonance SpectroscopyHyperpolarized 13C magnetic resonance spectroscopy (MRS) provides a unique opportunity to image reaction kinetics and enzyme activities in vivo. In contrast to conventional MRI, hy-perpolarization provides the ~50,000 fold increased signal-to-noise ratio needed to image both the injected substrate and downstream products, allowing real-time interrogation of mul-tiple key metabolic pathways. We have recently acquired the specialized equipment (Spinlab polarizer, GE Heathcare) to conduct both preclinical and clinical experiments, and while multiple 13C-labeled substrates have been studied in animal models, 13C-labeled pyruvate is the first of these to reach clini-cal trials. Located at a crucial hub in glucose metabolism, py-ruvate and its products provide key information for assessing the important balance among tissue energy needs, raw mate-rial requirements for cell division, and oxygen availability. Fu-ture studies include the assessment of oxidative phosphoryla-tion, and other key metabolic pathways, optimized mapping of 1H metabolite distributions throughout the body, and quantify-ing neurotransmitter levels and cycling rates in the brain.

Digital PET/CTThe world’s first digital PET/CT scanner, the Discovery MI manufactured by GE, was installed in the Nuclear Medicine and Molecular Imaging Clinic at Stanford Healthcare in August 2016. The digital detector design, extended axial Field of View (FOV), combined with Time-of-Flight capabilities and Q-Clear reconstruction software result in significant improvements in sensitivity and resolution for a direct impact on scan times, administered dose levels, and detection of small lesions. The Discovery MI PET/CT camera is used to image both clinical patients and research participants with the goal of achieving optimal patient care in a wide range of promising applications such as oncology, cardiology, and neurology.

Digital PET/MRPET/MR is a two-in-one test that combines images from a positron emission tomography (PET) scan and a magnetic resonance imaging (MRI) scan. This new hybrid technology harnesses the strengths of PET and MRI to produce some of the most highly detailed pictures of the inside of the hu-man body currently available. Doctors use those pictures to diagnose medical conditions and plan their treatment. For ex-ample, PET/MRI scans of the brain are useful in the care of Alzheimer’s disease, epilepsy, and brain tumors.

Stanford has two SIGNA PET/MRI scanners installed, one in the Lucas Center dedicated to pre-clinical research stud-ies and one in the Stanford Neuroscience Health Center for clinical imaging. A third system was installed at the new Lucile Packard Children’s Hospital.

Page 19: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

MIPS Collaborators

http://mips.stanford.edu

Ben and Catherine Ivy FoundationCanary FoundationChambers Family FoundationChild Health Research Fund, Stanford UniversityChild Health Research Institute at StanfordCLARIONS FoundationDamon Runyon Cancer Research FoundationDesmoid Tumor Research FoundationGoogleMusculoskeletal Transplant FoundationPeter Michael FoundationKenneth Rainin FoundationVarian Medical Systems FoundationXygen Diagnostics, Inc.

American College of Radiology Boston Children’s HospitalCalifornia Cancer ConsortiumThe Catholic University of Korea, SeoulChinese Academy of SciencesCopenhagen University Hospital Cornell UniversityDartmouth College David Geffen School of Medicine at UCLAEmory UniversityFondazione Bruno KesslerHarvard University Institut Lumière MatièreInternational Science and Technology CenterJohns Hopkins UniversityLeiden University Medical CenterMassachusetts General HospitalMayo ClinicMcGill UniversityMemorial-Sloan Kettering Cancer CenterMichigan State UniversityMITNational Institute of Advanced Industrial Science and Technology (AIST), JapanNational Institute of Radiological Sciences (NIRS), JapanNCI Experimental Therapeutics Clinical Trials Net-work (ETCTN)Norwegian Medical Cyclotron Centre Oregon Health & Science UniversityOslo University HospitalSRM University, IndiaStanford Neurosciences InstituteTechnical University of MunichThe University of Texas MD Anderson Cancer Cen-terThe Second Affiliated Hospital of Harbin Medical University, ChinaThe University of Alabama at Birmingham UCLAUCSFUniversity Medical Center, UtrechtUniversity of ArizonaUniversity of BradfordUniversity of BrisbaneUniversity of California at Berkeley

ActiveAdvanSiDAmplexi-LLCAvelas BiosciencesBayerBell BiosystemsBraccoBristol-Myers SquibbBruker BiospinCareviveCellSight TechnologiesCorvus PharmaceuticalsDynavaxFinisarGEGenentechGoogleGround Flour Pharmaceuticals, Inc.HamamatsuIntelInviCROJounce TherapeuticsKaryopharm TherapeuticsKETEK GmbHLI-COR BiosciencesLoxo OncologyMagnetic InsightMolecular Targeting Technologies, (MTTI), Inc.

Gift Giving Foundations

InstitutionsIndustryMR SolutionsNovadaq Technologies, Inc.NovartisNuvOx PharmaOcean NanotechOlympusOncoNano MedicinePfizerPhilipsPIMODPiramal ImagingPliant TherapeuticsPrecision X-RayR&A, FranceRadiation Monitoring DevicesReCor MedicalSci-Engi-Medco Solutions, Inc.SensLSiemensSiteOne Therapeutics, IncSOFIESTMicroelectronicsStryker EndoscopySurgiMabTransDerm, Inc.Varian Medical SystemsZiteo MedicalZurich MedTech

University of California at DavisUniversity of California at IrvineUniversity of CambridgeUniversity of ChicagoUniversity of CopenhagenUniversity of Duisburg-EssenUniversity of GenevaUniversity of GenoaUniversity of HawaiiHeidelberg UniversityUniversity of KentuckyUniversity of MainzUniversity of MichiganUniversity of Mississippi University of North Carolina at Chapel HillSanta Clara UniversityUniversity of Southern CaliforniaUniversity of TorontoUniversity of UtahUniversity of VirginiaUniversity of WashingtonWest Virginia University (WVU)Univversity of ZurichVilnius UniversityWashington State UniversityWestern UniversityYale University

Page 20: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

MIPS World Map

http://mips.stanford.edu

Previous Members by City

28Number of MIPS

Faculty

34Number of

Affiliated MIPS Faculty

503Number of MIPS

Alumni

7Number of Affiliated

Departments

207Number of Trainees

Page 21: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Recent Highlights

http://mips.stanford.edu

February 2013

• MIPS-Stanford Collaboration Leads to a Novel Bioprobe

March 2013

• Dr. Amelie Lutz Received First-Time Genitourinary Paper Presenter Award

• Dr. Sam Gambhir’s Research on Imaging Therapeutic Stem Cells Received Media Coverage

April 2013

• Jelena Levi’s Photoacoustic Imaging Research Highlighted in Clinical Cancer Research

• Dr. Adam de la Zerda Featured in Nature

May 2013

• Dr. Chris Contag’s Research on Tracking Metastatic Cancer Cell Interactions In Vivo Received Media Attention

• Jongho Jeon and Bin Shen are 20th International Sympo-sium on Radiopharmaceutical Sciences Award Recipients

• Huaijun (Morgan) Wang, Steven Machtaler, and Sunitha Bachawal received 2013 Korean Society of Ultrasound in Medicine Awards

June 2013

• Julie Kulm and Bin Shen Received the 2013 Society of Nu-clear Medicine and Molecular Imaging Award

• Molecular Imaging Primer Electronic Textbook written by Dr. Sam Gambhir is available online

July 2013

• Raiyan Zaman Awarded AHA Fellowship• Dr. Craig Levin’s Collaboration Publishes a New Article in

PNAS• Dr. Heike Daldrup-Link’s Research on Tracking Transplanted

Stem Cells Receives Extensive Media Coverage• Michelle James Receives Travel Fellwoship from AAIC

August 2013

• Saeed Mohammadi Received 2013 BioNanotechnology Summer Institute Best Poster Award

September 2013

• Dr. Jianghong Rao’s and Dr. Fred Chin’s Research Featured on the Cover of the Journal Angewandte Chemie

October 2013

• MIPS Interdisciplinary Team Creates Smart Nanoparticles for Cancer Therapy

• Sunitha Bachawal, Sarah Bohndiek, Colin Carpenter, Carmel Chan, Fanny Chapelin, Kai Cheng, Christopher Dove, Aileen Hoehne, Ohad Ilovich, Michelle James, Jesse Jokerst, Chris-topher Klenk, Ferdinand Knieling, Frederick Lartey, Steven Machtaler, Hossein Nejadnik, Mikael Palner, Kanyi Pu, John Ronald, Laura Sasportas, Bryan Smith, Sui-Seng Tee, Thillai Veerapazham, Huaijun Wang, Tzu Yin Wang, Katheryne Wilson, Deju Ye, and Raiyan Zaman received 2013 World Molecular Im-aging Congress Travel and Poster Awards

• Mikael Palner Nominated for 2013 World Molecular Imaging Congress Young Investigator Award

• Khun Visith Kue Received 2013 World Imaging Congress Young Investigator Award

November 2013

• “Laboratory Protocols” Electronic Textbook by Fanny Chapelin, Graham Beck, Olga Lenkov and Dr. Heike Daldrup-Link is Made Available Online

• Dr. Avnesh Thakor’s and Dr. Sam Gambhir’s Research Fea-tured on CA: A Cancer Journal for Clinicians Cover

• Dr. Sam Gambhir’s Research Featured on NanoLetters Cover

December 2013

• Fanny Chapelin Received France “Best Engineer of the Year” Award

Janaury 2014

• Dr. Adam de la Zerda Elected to Forbes Magazine “30 Under 30”

February 2014

• Dr. Heike Daldrup-Link’s and Dr. Jianghong Rao’s Research Featured on Cover of Small

• New MRI Technique by Dr. Heike Daldrup-Link and Dr. Michael Moseley Could Offer Radiation-free Alternative for Visualising Cancerous Tumours in Children

March 2014

• MIPS Research Featured in the Journal of the American Chemi-cal Society Spotlight

April 2014

• Dr. Sandip Biswal’s and Dr. Fred Chin’s Research Highlighted in Chemical & Engineering News Cover Story

• Michelle James Received First Prize at INMiND TSPO Sympo-sium

• Dr. Sam Gambhir’s Research Identifies More Efficient Cancer Drug Delivery Method in Nature Nanotechnology

May 2014

• Ophir Vermesh Awarded 2014 Dean’s Fellowship

June 2014

• Laura Sasportas Received the Young Investigator Award at 2014 SNMMI

August 2014

• Dan Feng Awarded Translational Research and Applied Medi-cine (TRAM) Grant

November 2014

• MIPS Collaborative Research Featured in the Journal of the American Chemical Society Spotlight and Nature SciBx

• Huaijun (Morgan) Wang Awarded RSNA 2014 Travel Grant• Katheryne Wilson Awarded Travel Grant from the Henzl-Gabor

Young Women in Science Fund • Dr. Sam Gambhir Named Fellow of American Association for the

Advancement of Science (AAAS)

December 2014

• Hossein Nejadnik Received 2014 RSNA Trainee Research Prize• Laura Sasportas Received France “Best Engineer of the Year”

Award• Michelle James Received Media Coverage for New Study Imag-

ing Alzheimer’s Disease Therapy Response - Research high-lighted in AlzForum and Neurology Today

• MIPS/Stanford Publishes First Clinical Paper Reporting the Use of Simultaneous PET/MRI with Time-of-flight Capability in Can-cer Patients

2014

2013

January 2015

• Maryam Aghighi Recevied 2015 NAIRS Research Award

February 2015

• Dr. Zhen Cheng’s Protein Microarrays for Studies in Biomark-ers and Post Translational Modification Paper highlighted by Advance Materials as the Inside Cover

March 2015

• Dr. Sam Gambhir’s Research Uses Tumor-activatable Minicir-cles for Early Detection of Cancer

• Dr. Heike Daldrup-Link Elected to the American Society for Clini-cal Investigation (ASCI)

• Dr. Jesse Jokert’s and Dr. Sam Gambhir’s Joint Research Fea-tured on the Cover of Theranostics

• Aaron Mayer and Surya Murty Awarded NSF Fellowships

April 2015

• Drs. Tim Witney and Michelle James Received SNM Alavi Man-dell Awards

May 2015

• Dr. Jesse Jokerst’s and Dr. Sam Gambhir’s Research Featured on the Inside Cover of Analyst

2015

Page 22: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Recent Highlights

http://mips.stanford.edu

June 2015

• Dr. Adam de la Zerda Selected as 2015 Pew-Stewart Schol-ar for Cancer Research

July 2015

• The Robarts Research Institute awards the 2015 J. Allyn Taylor International Prize in Medicine to Dr. Sam Gambhir

September 2015

• Dr. Zhen Cheng’s Research Featured as the Inside Cover in Advanced Materials

October 2015

• Drs. Meghdad Garmestani and Saeid Zanganeh are 2015 ISMRM Travel Award Recipients

• Dr. Sam Gambhir’s Research Featured on the Cover of the Journal Science Translational Medicine

December 2015

• Dr. Anne Muehe Received 2015 RSNA Trainee Research Prize

• Dr. Sam Gambhir Named National Academy of Inventors Fellow

• Dr. Zhen Cheng’s Collaborative Research Publishes First Clinical Translatable NIR-II Fluorescent Dye

January 2016

• Dr. Sam Gambhir’s Research Featured on the Cover of the Journal of Neuro-Oncology

• Dr. Kai Li Received 2016 BIT Forum Travel Award

March 2016

• Dr. Michelle James Wins MRC Suffrage Science Award• Dr. Jessica Klockow Received American Chemical Society

CIBA/YCC Young Scientist Award• Dr. Adam de la Zerda’s Research Develops New Imaging

Technique: MOZART

April 2016

• Dr. Michelle James Received JNM Editors’ Choice Award

• Dr. Lawrence Fung Received NeuroReceptor Mapping 2016 Young Investigator Award

June 2016

• Dr. Hamed Arami Selected to Present at the University of Wash-ington’s 6th Annual Distinguished Young Scholars Seminar

• Dr. Hao Chen Received SNMMI - CMIIT 2016 Young Investiga-tor Award

• Dr. Joseph Wu’s Stem Cell Research is Highlited in OZY under the section “Rising Stars”

July 2016

• Dr. Adam de la Zerda Presents at 2016 TedX Stanford• MOU Initiated by MIPS Researchers Signed Between Stanford

University and Catholic University of Korea

September 2016

• WMIC 2016 Award Recipients. Travel Stipends: Ahmed El Kaf-fas, Kai Li, Orly Liba, Michael Mustanduno, Eliott SoRelle, Nutte Teraphongphom, Huaijun Wang. Poster Award Finalists: Ahmed El Kaffas, Sharon Hori, Kai Li, Michael Mustanduno. Women in Molecular Imaging Network Scholar Awards: Orly Liba, Nutte Teraphongphom.

October 2016

• Dr. Heike Daldrup-Link’s Research Published in Nature Nano-technology

November 2016

• Dr. Chris Contag is Named Recipient of the 2017 SPIE Britton Chance Biomedical Optics Award for Bioluminescence Imaging

• Chen-Ming Chang, PhD Student, Received IEEE 2016 Best Oral Presentation Student Award

• Li Tao was awarded the Stanford Graduate Fellowship in Sci-ence and Engineering (SGF)

December 2016

• Dr. Ophir Vermesh Received the SURPAS Best Talk Award.• Dr. Dan Spielman was Named a Fellow of ISMRM• MIPS Researchers Present a Blood Test for a Better Way to

Manage Lung Cancer• Ophir Vermash wins Best Poster Award at IEEE Micro and Nan-

otechnology in Medicine

Janaury 2017• Scarlett Guo receives Bio-X USRP Research Fellowship• Study of the First Human Reporter Gene Imaging for Cancer

Immunotherapy is Published by MIPS Researchers• The Inagural Molecular Imaging Young Investigator (MIYI) Prize

is Awarded to Drs. Katheryne Wilson and Jinghang Xie

February 2017

• Dr. Adam de la Zerda is Named a Chan Zuckerberg Biohub Ju-nior Investigator

• Dr. Joseph Wu Develops a Test that can Screen for Cardiotoxic-ity in new Chemotherapy Drugs

• MIPS Collaboration with Chemistry Department Leads to New Functional Delivery System of mRNA

March 2017

• Ophir Vermesh Received Poster Award at the Bio-X Interdisci-plinary Initiatives Seed Grants Symposium

April 2017

• Dr. Juergen Willmann and Dr. Sanjiv Sam Gambhir Publish the First-in-human Ultrasound Molecular Imaging in Patients with Breast and Ovarian Lesions

May 2017

• Dr. Joseph Wu Received the American Heart Association Merit Award

• Amin Aalipour Named 2017 Paul & Daisy Soros Fellow

June 2017

• Dr. Raiyan Zaman Received 2017 SNMMI Henry N Wagner Jr, MD, Best Paper Award

• Dr. Juergen Willmann received the 2017 Academy for Radiol-ogy & Biomedical Imaging Research Distinguished Investigator Award

• Hao Chen received SNMMI-CMIIT 2017 Young Investigator Award

July 2017

• Stanford Launches Project Baseline Study by Enrolling First Participant

August 2017

• Dr. Heike Daldrup-Link is Appointed Director of Pediatric Mo-lecular Imaging

• Dr. Raag Airan is the Finalist for the Science-PINS Prize for Neuromodulation

• Drs. Li Tao and Joshua Cates Receive the 2017 Valentin T. Jordanov Radiation Instrumentation Travel Grant to Participate at the 2017 IEEE NSS-MIC

September 2017

• Dr. Sanjiv Sam Gambhir is Appointed President of the IS3R• WMIC 2017 Award Recipients. Young Investigator Award: Pat-

rick McCormick. Young Investigator Award Finalist: Suchismita Mohanty. Best Poster Award: Emily Johnson, PhD. WIMIN Trav-el Awards: Louise Kiru, Suchismita Mohanty, Nutte Teraphong-phom, Nynke van den Berg

October 2017

• MIPS Researchers Develop New PET Tracer that can Identify Most Bacterial Infections

November 2017

• MIPS Research on Noninvasive Neuromodulation with Nanopar-ticles using FUS Highlighted in Nature

December 2017

• Drs. Daldrup-Link and Rao Elected to AIMBE College of Fellows2016

2017

Page 23: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Recent Publications

http://mips.stanford.edu

Abbaszadeh S, Levin CS. New-generation small animal positron emission tomography system for molecular imaging. Journal of Medical Imaging (Bellingham). 2017; 4(1): 011008.

Aghighi M, Boe J, Rosenberg J, Von Eyben R, Gawande RS, Petit P, Sethi TK, Sharib J, Marina NM, DuBois SG, Dal-drup-Link HE. Three-dimensional Radiologic Assessment of Chemotherapy Response in Ewing Sarcoma Can Be Used to Predict Clinical Outcome. Radiology. 2016; 280(3):905-15.

Airan RD, Meyer RA, Ellens NP, Rhodes KR, Farahani K, Pomper MG, Kadam SD, Green JJ. Noninvasive Target-ed Transcranial Neuromodulation via Focused Ultrasound Gated Drug Release from Nanoemulsions. Nano letters. 2017; 17 (2): 652-659.

Ali R, Apte S, Vilalta M, Subbarayan M, Miao Z, Chin FT, Graves EE. 18F-EF5 PET Is Predictive of Response to Frac-tionated Radiotherapy in Preclinical Tumor Models. PLoS One. 2015; 10(10): e0139425.

Ananta JS, Paulmurugan R, Massoud TF. Tailored Nanoparticle Codelivery of antimiR-21 and antimiR-10b Augments Glioblastoma Cell Kill by Temozolomide: Toward a “Personalized” Anti-microRNA Therapy. Molecular Pharmaceutics. 2016; 13(9), 3164-3175.

Ananta JS, Paulmurugan R, Massoud TF. Temozolomide-loaded PLGA nanoparticles to treat glioblastoma cells: A bio-physical and cell culture evaluation. Neurological Research. 2016; 38: 51-9.

Antaris AL, Chen H, Cheng K, Sun Y, Hong G, Qu C, Diao S, Deng Z, Hu X, Zhang B, Zhang X, Yaghi OK, Alamparambil ZR, Hong X*, Cheng Z*, Dai H*. A small-molecule dye for NIR-II imaging. Nature Materials. 2016; 15(2): 235-42.

Antaris AL, Chen H, Diao S, Ma Z, Zhang Z, Zhu S, Wang J, Lozano AX, Fan Q, Chew L, Zhu M, Cheng K, Hong X, Dai H, Cheng Z. A high quantum yield molecule-protein complex fluorophore for near-infrared II imaging. Nature Com-munications. 2017; 8: 15269.

Bachawal SV, Jensen KC, Wison KE, Tian L, Lutz AM, Willmann JK. Breast cancer detection by B7-H3 targeted ultra-sound molecular imaging. Cancer Research. 2015; 75(12): 2501-9.

Bazalova-Carter M, Weil MD, Breitkreutz DY, Wilfley BP, Graves EE. Feasibility of external beam radiation therapy to deep-seated targets with kilovoltage x-rays. Medical physics.2017; 44(2): 597-607.

Bieniosek, MF, Cates JW, Levin CS. A multiplexed TOF and DOI capable PET detector using a binary position sensitive network. Physics in medicine and biology. 2016; 61(21): 7639-7651.

Bradford D, Reilly KM, Widemann BC, Sandler A, Kummar S. Developing therapies for rare tumors: opportunities, chal-lenges and progress. Expert Opinion on Orphan Drugs. 2016; 4(1): 93-103.

Burridge PW, Li YF, Matsa E, Wu H, Ong S, Sharma A, Holmstrom A, Chang AC, Coronado MJ, Ebert AD, Knowles JW, Telli ML, Witteles R M, Blau HM, Bernstein D, Altman RB, Wu JC. Human induced pluripotent stem cell-derived cardio-myocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nature Medicine. 2016; 22 (5): 547-556.

Burridge PW, Li YF, Matsa E, Wu H, Ong SG, Sharma A, Holmstrom A, Chang AC, Coronado MJ, Ebert AD, Knowles JW, Telli ML, Witteles RM, Blau HM, Bernstein D, Altman RB, Wu JC. Human induced pluripotent stem-derived cardio-myocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nature Medicine. 2016; 22(5): 547-56.

Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Guetgemann I, Eilers M, Felsher DW. MYC regulates the antitumor immune response through CD47 and PD-L1. Science. 2016; 352 (6282): 227-231.

Cui L, Rao J. Semiconducting polymer nanoparticles as photoacoustic molecular imaging probes. Nano-medicine and Nanobiotechnology. 2016; 9:e1418.

Dedic A, Lubbers MM, Schaap J, Lammers J, Lamfers EJ, Rensing BJ, Braam RL, Nathoe HM, Post JC, Nielen T, Beelen D, le Cocq d’Armandville MC, Rood PP, Schultz CJ, Moelker A, Ouhlous M, Boersma E, Nieman K. Coronary CT Angiography for Suspected ACS in the Era of High-Sensitivity Troponins: Ran-domized Multicenter Study. J Am Coll Cardiol. 2016 Jan 5;67(1):16-26.

Devulapally R, Sekar NM, Sekar TV, Foygel K, Massoud TF, Willmann JK, Paulmurugan R. Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative breast can-cer therapy. ACS Nano. 2015; 9(3): 2290-302.

Do K, Wilsker D, Ji J, Zlott J, Freshwater T, Kinders RJ, Collins J, Chen AP, Doroshow JH, Kummar S. Phase I Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients with Refractory Solid Tumors. Journal of Clinical Oncology. 2015; 33(30): 3409-15.

Doroshow JH, Kummar S. Translational research in oncology-10 years of progress and future prospects. Nature Reviews Clinical Oncology. 2014; 11(11): 649-62.

Grant AM, Deller TW, Khalighi MM, Maramraju, SH, Delso G, Levin CS. NEMA NU 2-2012 performance studies for the SiPM-Based Time-of-Flight PET component of the GE SIGNA PET/MR System. Medical Physics. 2016; 43: 2334.

Hamilton AM, Aidoudi-Ahmed S, Sharma S, Kotamraju VR, Foster PJ, Sugahara KN, Ruoslahti E, Rutt BK. Nanoparticles coated with the tumor-penetrating peptide iRGD reduce experimental breast cancer metas-tasis in the brain. Journal of Molecular Medicine (Berlin). 2015; 93(9): 991-1001.

Hardy JW, Levashova Z, Schmidt TL, Contag CH, Blankenberg FG. [99mTc]Annexin V-128 SPECT Moni-toring of Splenic and Disseminated Listeriosis in Mice: a Model of Imaging Sepsis. Molecular Imaging and Biology. 2015; 17(3): 345-54.

Holdsworth SJ, Rahimi MS, Ni WW, Zaharchuk G, Moseley ME. Amplified magnetic resonance imaging (aMRI). Magnetic resonance in medicine. 2016; 75(6): 2245-2254.

Ibragimov B, Xing L. Segmentation of organs-at-risks in head and neck CT images using convolutional neural networks. Medical Physics. 2017; 44(2), 547-557.

James ML, Belichenko N, Nguyen TV, Andrews L, Liu H, Bodapati D, Shen B, Cheng Z, Gambhir SS, Longo FM, Chin FT. Multimodality molecular imaging of translocator protein (18kDa) in a mouse model of Alzheimer’s disease using [18F]PBR06. Journal of Nuclear Medicine. 2015; 56(2): 311-16.

James ML, Belichenko NP, Nguyen TV, Andrews LE, Ding Z, Liu H, Bodapati D, Arksey N, Shen B, Cheng Z, Wyss-Coray T, Gambhir SS, Longo FM, Chin FT. PET Imaging of Translocator Protein (18 kDa) in a Mouse Model of Alzheimer’s Disease Using N-(2,5-Dimethoxybenzyl)-2-18F-Fluoro-N-(2-Phenoxyphenyl)Acetamide. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2015; 56 (2): 311-316.

James ML, Belichenko NP, Shuhendler AJ, Hoehne A, Andrews LE, Condon C, Nguyen TV, Reiser V, Jones P, Trigg W, Rao J, Gambhir SS, Longo FM. [F-18]GE-180 PET Detects Reduced Microglia Activation After LM11A-31 Therapy in a Mouse Model of Alzheimer’s Disease. Theranostics. 2017; 7 (6): 1422-1436.

King M, Carpenter C, Sun C, Ma X, Le Q, Sunwoo J, Cheng Z, MD; Pratx G, Xing L. A comparative evalu-ation of CCD-based beta imaging and Cerenkov luminescence imaging for FDG-guided surgery. Journal of

Page 24: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Recent Publications

http://mips.stanford.edu

Nuclear Medicine. 2015; 56: 1458-64.

King MT, Jenkins CH, Sun C, Carpenter CM, Ma X, Cheng K, Le QT, Sunwoo JB, Cheng Z, Pratx G, Xing L. Flexible radioluminescence imaging for FDG-guided surgery. Medical Physics. 2016; 43(1): 5298-5306.

Kodo K, Ong S, Jahanbani F, Termglinchan V, Hirono K, Inanloorahatloo K, Ebert AD, Shukla P, Abilez OJ, Churko JM, Karakikes I, Jung G, Ichida F, Wu SM, Snyder MP, Bernstein D, Wu JC. iPSC-derived cardiomyocytes reveal abnormal TGF-ß signalling in left ventricular non-compaction cardiomyopathy. Nature cell biology. 2016; 18(10): 1031-1042.

Kuchcinski G, Munsch F, Lopes R, Bigourdan A, Su J, Sagnier S, Renou P, Pruvo J, Rutt BK, Dousset V, Sibon I, Tour-dias T. Thalamic alterations remote to infarct appear as focal iron accumulation and impact clinical outcome. Brain. 2017; 140(7): 1931-1946.

Li Y, Choi PS, Casey SC, Dill DL, Felsher DW. MYC through miR-17-92 Suppresses Specific Target Genes to Maintain Survival, Autonomous Proliferation, and a Neoplastic State. Cancer cell. 2014; 26 (2): 262-272.

Liba O, de la Zerda A. Photoacoustic tomography: Breathtaking whole-body imaging. Nature Biomedical Engineering. 2017: 1(5) pp: 0075.

Liba O, Lew MD, SoRelle ED, Dutta R, Sen D, Moshfeghi DM, Chu S, de la Zerda A. Speckle modulating optical coher-ence tomography in living mice and humans. Nature Communications. 2017; 8: 15845.

Liba O, SoRelle E, Sen D, de la Zerda A. Contrast-enhanced optical coherence tomography with picomolar sensitivity for functional in vivo imaging. Scientific Reports. 2016; 18(6): 23337.

Loening AM, Litwiller DV, Saranathan M, Vasanawala SS. Increased speed and image quality for pelvic single shot fast spin echo imaging via variable refocusing flip angles and full-Fourier acquisition. Radiology. 2017; 282 (2): 561-568.

Loening AM, Saranathan M, Ruangwattanapaisarn N, Litwiller DV, Shimakawa A, Vasanawala SS. Increased speed and image quality in single shot fast spin echo imaging via variable refocusing flip angles. Journal of Magnetic Resonance Imaging. 2015; 42(6): 1747-1758.

Lubbers M, Dedic A, Coenen A, Galema T, Akkerhuis J, Bruning T, Krenning B, Musters P, Ouhlous M, Liem A, Niezen A, Hunink M, de Feijter P, Nieman K. Calcium imaging and selective computed tomography angiography in comparison to functional testing for suspected coronary artery disease: the multicentre, randomized CRESCENT trial. Eur Heart J. 2016 Apr 14;37(15):1232-43.

Minamimoto R, Hancock S, Schneider B, Chin FT, Jamali M, Loening A, Vasanawala S, Gambhir SS, Iagaru A. Pilot Comparison of Ga-68-RM2 PET and Ga-68-PSMA-11 PET in Patients with Biochemically Recurrent Prostate Cancer. Journal of Nuclear Medicine. 2016; 57(4): 557-562.

Minamimoto R, Loening A, Jamali M, Barkhodari A, Mosci C, Jackson T, Obara P, Taviani V, Gambhir SS, Vasanawala S, Iagaru AH. Prospective comparison of 99mTc MDP scintigraphy, combined 18F-NaF and 18F-FDG PET/CT and whole-body MRI in patients with breast and prostate cancers. Journal of Nuclear Medicine. 2015; 56(12): 1862-68.

Mullick Chowdhury S, Wang TY, Bachawal S, Devulapally R, Choe JW, Abou Elkacem L, Khuri Yakub B, Wang DS, Tian L, Paulmurugan R, Willmann JK. Ultrasound-Guided Therapeutic Modulation of Hepatocellular Carcinoma using Complementary microRNAs. J of Controlled Release 2016; Sep 28;238:272-80.

Natarajan A, Türkcan S, Gambhir SS, Pratx G. A multiscale framework for imaging radiolabeled therapeutics. Molecular Pharmaceuticals. 2015; 12(12): 4554-4560.

Nejadnik H, Ye D, Lenkov O, Donig J, Martin J, Castillo R, Derugin N, Sennino B, Rao J, Daldrup-Link H. MR imaging of

stem cell apoptosis in arthritic joints with a novel, caspase-activatable contrast agent for MR imaging. ACS Nano. 2015; 9(2): 1150-60.

Parashurama N, Ahn B-C, Ziv K, Ito K, Paulmurugan R, Willmann JK, Chung J, Ikeno F, Swanson JC, Merk DR, Lyons JK, Yerushalmi D, Teramoto T, Kosuge H, Dao CN, Ray P, Patel M, Chang Y-F, Mahmoudi M, Gambhir SS. Multimodality molecular imaging of cardiac cell transplantation: Part I. Reporter gene design, characterization, and optical in vivo imaging of bone marrow stromal cells after myocardial infarction. Ra-diology. 2016; 280(3), 815-825.

Parashurama N, Ahn B-C, Ziv K, Ito K, Paulmurugan R, Willmann JK, Chung J, Ikeno F, Swanson JC, Merk DR, Lyons JK, Yerushalmi D, Teramoto T, Kosuge H, Dao CN, Ray P, Patel M, Chang Y-F, Mahmoudi M, Gambhir SS. Multimodality molecular imaging of cardiac cell transplantation: Part II. in vivo imaging of bone marrow stromal cells in swine with PET/CT and MR imaging. Radiology. 2016; 280(3), 826-836.

Park JM, Khemtong C, Liu SC, Hurd RE, Spielman DM. In vivo assessment of intracellular redox state in rat liver using hyperpolarized [1-13 C]Alanine. Magn Reson Med. 2017 May;77(5):1741-1748.

Park JM, Wu M, Datta K, Liu SC, Castillo A, Lough H, Spielman DM, Billingsley KL. Hyperpolarized sodium [1-13C]-Glycerate as a probe for assessing glycolysis in vivo. J Am Chem Soc. 2017; 139(19):6629-6634.

Park S-M, Aalipour A, Vermesh O, Yu H, Gambhir SS. Towards clinically translatable in vivo nanodiagnos-tics. Nature Reviews Materials. 2017; 2:17014.

Park S-M, Wong DJ, Ooi CC, Kurtz DM, Vermesh O, Aalipour A, Suh S, Pian KL, Chabon JJ, Lee SH, Ja-mali M, Say C, Carter JN, Lee LP, Kuschner WG, Schwartz EJ, Shrager JB, Neal JW, Wakelee HA, Diehn M, Nair VS, Wang SX, Gambhir SS. Molecular profiling of single circulating tumor cells from lung cancer patients. Proceedings of the National Academy of Sciences of the United States of America. 2016; 113(52), E8379-E8386.

Paulmurugan R, Bhethanabotla R, Mishra K, Devulapally R, Foygel K, Sekar TV, Ananta JS, Massoud TF, Joy A. Folate Receptor-Targeted Polymeric Micellar Nanocarriers for Delivery of Orlistat as a Repurposed Drug against Triple-Negative Breast Cancer. Molecular Cancer Therapeutics. 2016; 15(2): 221-31.

Pu K, Chattopadhyay N, Rao J. Recent advances of semiconducting polymer nanoparticles in in vivo mo-lecular imaging. Journal of Controlled Release. 2016; 240, 312-322.

Ronald JA, Chuang HY, Dragulescu-Andrasi A, Hori SS, Gambhir SS. Detecting Cancers Through Tumor-Activatable Minicircles That Lead to a Detectable Blood Biomarker. Proceedings of the National Academy of Sciences (USA). 2015; 112(10): 3068-73.

Rosenthal E, Moore L, Tipirneni K, de Boer E, Stevens TM, Hartman YE, Carroll WR, Zinn KR, Warram JM. Sensitivity and Specificity of Cetuximab-IRDye800CW to Identify Regional Metastatic Disease in Head and Neck Cancer. Clinical Cancer Research. 2017; 23(16): 4744-4752.

Rosenthal EL, Chung TK, Parker WB, Allan PW, Clemons L, Lowman D, Hong J, Hunt FR, Richman J, Conry RM, Mannion K, Carroll WR, Nabell L, Sorscher EJ. Phase I dose-escalating trial of Escherichia coli purine nucleoside phosphorylase and fludarabine gene therapy for advanced solid tumors. Annals of Oncology. 2015; 26(7): 1481-7.

Rusckowski M, Wang Y, Blankenberg FG, Levashova Z, Backer MV, Backer JM. Targeted scVEGF/Lu-177 radiopharmaceutical inhibits growth of metastases and can be effectively combined with chemothera-py. EJNMMI Research. 2016; 6(4): 1-9.

Page 25: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Recent Publications

http://mips.stanford.edu

Sekar TV, Foygel K, Devulapally R, Kumar V, Malhotra SV, Massoud TF, Paulmurugan P. Molecular imaging biosensor monitors p53 Sumoylation in cells and living mice. Analytical Chemistry, 2016; 88, 11420-11428.

Sekar TV, Foygel K, Massoud TF, Gambhir SS, Paulmurugan R. A transgenic mouse model expressing an ERα folding biosensor reveals the effects of Bisphenol A on estrogen receptor signaling. Scientific Reports. 2016; 6: 34788.

Sengupta D, Miller S, Marton Z, Chin F, Nagarkar V, Pratx G. Bright Lu2O3:Eu Thin-Film Scintillators for High-Resolution Radioluminescence Microscopy. Advanced Healthcare Materials. 2015; 4(14): 2064-70.

Sengupta D, Pratx G. Single-cell characterization of 18F-FLT uptake with radioluminescence microscopy. Journal of Nuclear Medicine. 2016; 57(7): 1136-40.

Sharma A, Burridge PW, McKeithan WL, Serrano R, Shukla P, Sayed N, Churko JM, Kitani T, Wu H, Holmström A, Matsa E, Zhang Y, Kumar A, Fan AC, Del Álamo JC, Wu SM, Moslehi JJ, Mercola M, Wu JC.High-throughput screen-ing of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Science translational medicine. 2017; 9(377): eaaf2584.

Sharma GVM, Kumar KS, Reddy SV, Nagalingam A, Cunningham KM, Ummanni R, Hugel H, Sharma D, Malhotra SV. Synthesis and Biological Evaluation of Triazole-Vanillin Molecular Hybrids as Anti-Cancer Agents. Current Bioactive Compounds. 2017,

Sheahan AV, Kai C, Sekar TV, Paulmurugan R, Massoud TF. A molecular imaging biosensor detects in vivo protein fold-ing and misfolding. Journal of Molecular Medicine. 2016; 94: 799-808.

Shen B, Behera D, James M, Reyes S, Cipriano P, Andrews L, Klukinov M, Lutz A, Mavlyutov T, Rosenberg J, Ruoho A, McCurdy C, Gambhir S, Yeomans D, Biswal S, Chin F. Visualizing Nerve Injury in a Neuropathic Pain Model with [18F]FTC-146 PET/MRI. Theranostics. 2017; 7(11), 2794-2805.

Shen B, Park JH, Hjørnevik T, Cipriano PW, Yoon D, Gulaka PK, Holly D, Behera D, Avery BA, Gambhir SS, McCurdy CR, Biswal S, Chin FT. Radiosynthesis and First-In-Human PET/MRI Evaluation with Clinical-Grade [18F]FTC-146. Molecular Imaging and Biology. 2017; 19(5), 779- 786.

Si P, Sen D, Dutta R, Yousefi S, Dalal R, Winetraub Y, Liba O, de la Zerda A. In Vivo Molecular Optical Coherence To-mography of Lymphatic Vessel Endothelial Hyaluronan Receptors. Scientific Reports. 2017; 7: 1086.

Smith BR, Gambhir SS. Nanomaterials for in Vivo Imaging. Chemical Reviews. 2017; 117(3), 901-986.

Soman S, Prasad G, Hitchner E, Massaband P, Moseley ME, Zhou W, Rosen AC. Brain structural connectivity distin-guishes patients at risk for cognitive decline after carotid interventions. Human Brain Mapping. 2016; 37(6): 2185-94.

Song X, Airan RD, Arifin DR, Bar-Shir A, Kadayakkara DK, Liu G, Gilad AA, van Zijl PC, McMahon MT, Bulte JW. Label-free in vivo molecular imaging of underglycosylated mucin-1 expression in tumour cells. Nature Communications. 2015; 6:6719.

SoRelle E, Liba O, Husssain Z, Gambhir M., de la Zerda A. Biofunctionalization of Large Gold Nanorods Realizes Ultrahigh-Sensitivity Optical Imaging Agents. Langmuir. 2015, 31(45): 12339-47.

Thakor AS, Gambhir SS. Nanooncology: The Future of Cancer Diagnosis and Therapy. CA: A Cancer Journal for Clini-cians. 2013; 63 (6): 395-418.

Thakor AS, Jokerst, JV, Ghanouni P, Campbell JL, Mittra E, Gambhir SS. Clinically Approved Nanoparticle Imaging Agents. Journal of Nuclear Medicine. 2016; 57(12), 1833-1837.

Thakor AS, Sangha BS, Ho SG, Warnock GL, Meloche M, Liu DM. Percutaneous autologous pancreatic islet cell transplantation for traumatic pancreatic injury. Journal of Clinical Endocrinology & Metabolism. 2015; 100(4): 1230-33.

Tipirneni KE, Warram JM, Moore LS, Prince AC, de Boer E, Jani AH, Wapnir IL, Liao JC, Bouvet M, Behnke NK, Hawn MT, Poultsides GA, Vahrmeijer AL, Carroll WR, Zinn KR, Rosenthal E. Oncologic Procedures Amenable to Fluorescence-guided Surgery. Annals of Surgery. 2017; 266(1): 36-47.

Tummers WS, Warram JM, Tipirneni KE, Fengler J, Jacobs P, Shankar L, Henderson L, Ballard B, Pogue BW, Weichert JP, Bouvet M, Sorger J, Contag CH, Frangioni JV, Tweedle MF, Basilion JP, Gambhir SS, Rosenthal EL. Regulatory aspects of optical methods and exogenous targets for cancer detection. Cancer Research. 2017; 77(9), 2197-2206.

Wang H, Dong P, Liu H, Xing L. Development of an autonomous treatment planning strategy for radiation therapy with effective use of population-based prior data. Medical Physics. 2017; 44, 389-396.

Wang TY, Choe JW, Pu K, Devulapally R, Bachawal S, Machtaler S, Chowdhury SM, Luong R, Tian L, Khuri-Yakub B, Rao J, Paulmurugan R, Willmann JK. Ultrasound-guided Delivery of microRNA Loaded Nanoparticles into Cancer. Journal of Controlled Release. 2015; 203: 99-108.

Wilson KE, Bachawal SV, Abou-Elkacem L, Jensen K, Machtaler S, Tian L, Willmann JK. Spectroscopic Photoacoustic Molecular Imaging of Breast Cancer using a B7-H3-targeted ICG Contrast Agent. Theranos-tics. 2017 Apr 3;7(6):1463-1476.

Winkler SA, Schmitt F, Landes H, DeBever J, Wade T, Alejski A, Rutt BK. Gradient and shim technologies for ultra high field MRI. NeuroImage. 2016; http://dx.doi.org/10.1016/j.neuroimage.2016.11.033.

Wu H, Lee J, Vincent JG, Wang Q, Gu W, Lan F, Churko J, Sallam K, Matsa E, Sharma A, Gold JD, Engler AJ, Xiang YK, Bers DM, Wu JC. Epigenetic regulation of phosphodiesterases 2A and 3A underlies com-promised β-adrenergic signaling in iPSC model of dilated cardiomyopathy. Cell Stem Cell. 2015; 17(1): 89-100.

Ye D, Shuhendler AJ, Cui L, Tong L, Tee SS, Tikhomirov G, Felsher D, Rao J. Bioorthogonal cyclization and in situ self-assembly of small-molecule probes for imaging caspase activity in living mice. Nature Chemis-try. 2014; 6: 519-26.

Zanganeh S, Hutter G, Spitler R, Lenkov O, Mahmoudi M, Shaw A, Pajarinen JS, Nejadnik H, Goodman S, Moseley M, Coussens LM, Daldrup-Link HE. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nature nanotechnology. 2016; 11 (11): 986-994.

Zhang R, Cheng K, Antaris AL, Ma X, Yang M, Ramakrishnan S, Liu G, Lu A, Dai H, Tian M, Cheng Z. Hybrid anisotropic nanostructures for dual-modal cancer imaging and image-guided chemo-thermo thera-pies. Biomaterials. 2016; 103: 265-277.

Zhou X, Cipriano P, Kim B, Dhatt H, Rosenberg J, Mittra E, Do B, Graves E, Biswal S. Detection of nocicep-tive-related metabolic activity in the spinal cord of low back pain patients using 18F-FDG PET/CT. Scandi-navian Journal of Pain. 2017. 15: 53-57.

Page 26: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

MIPS Events

http://mips.stanford.edu

From top: MIPS Retreat; Farewell party; Team MIPS for the Canary Challenge; MIPS Retreat; MIPS Retreat.

Clockwise from top left: 2017 Radiology Joint Research Retreat; 2016 MIPS Retreat; 2016 MIPS Retreat; 2016 MIPS Retreat; 2017 WMIC MIPS Reception; 2017 WMIC MIPS Reception; Lab party; Lab BBQ; 2016 MIPS Retreat.

Page 27: Welcome [med.stanford.edu]med.stanford.edu/.../aboutus/2017-MIPS-brochure.pdf · via sound (ultrasound, photoacoustic), magnetism (MRI or magnetic resonance imaging, MPI or magnetic

Directions to the James Clark Center (home of MIPS)From Bayshore US Highway 101 North or SouthExit on University Ave. South/West. Proceed for several miles on University Ave. University Ave. becomes Palm Drive after you pass El Camino Real. Take Palm Drive past Arboretum Road. Then turn right on Campus Drive. Make a left at Roth Way and another left into the parking lot. Meters operate until 4:00PM.

From 280 North or SouthExit Sand Hill Road East. Follow this road for several miles. Take a right on Stock Farm Road and then turn left on Campus Drive. At Roth Way, make a right turn and then a left into the parking lot. Meters operate until 4:00PM.

The James H. Clark Center318 Campus DriveEast Wing, First FloorStanford, CA 94305-5427

Tel: (650) 724-1407http://mips.stanford.edu/