Network


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

Hotspot


Dive into the research topics where Susanne Kossatz is active.

Publication


Featured researches published by Susanne Kossatz.


Scientific Reports | 2016

Detection and delineation of oral cancer with a PARP1 targeted optical imaging agent.

Susanne Kossatz; Christian Brand; Stanley Gutiontov; Jonathan T. C. Liu; Nancy Y. Lee; Mithat Gonen; Wolfgang A. Weber; Thomas Reiner

Earlier and more accurate detection of oral squamous cell carcinoma (OSCC) is essential to improve the prognosis of patients and to reduce the morbidity of surgical therapy. Here, we demonstrate that the nuclear enzyme Poly(ADP-ribose)Polymerase 1 (PARP1) is a promising target for optical imaging of OSCC with the fluorescent dye PARPi-FL. In patient-derived OSCC specimens, PARP1 expression was increased 7.8 ± 2.6-fold when compared to normal tissue. Intravenous injection of PARPi-FL allowed for high contrast in vivo imaging of human OSCC models in mice with a surgical fluorescence stereoscope and high-resolution imaging systems. The emitted signal was specific for PARP1 expression and, most importantly, PARPi-FL can be used as a topical imaging agent, spatially resolving the orthotopic tongue tumors in vivo. Collectively, our results suggest that PARP1 imaging with PARPi-FL can enhance the detection of oral cancer, serve as a screening tool and help to guide surgical resections.


The Journal of Nuclear Medicine | 2017

Molecular Imaging of PARP

Thomas Reiner; Brandon Carney; Susanne Kossatz

The poly(adenosine diphosphate–ribose)polymerase (PARP) family of enzymes is an important factor in the cellular DNA damage response and has gained much attention for its role in many diseases, particularly cancer. Targeted molecular imaging of PARP using fluorescent or radiolabeled tags has followed on the success of therapeutic inhibitors and gained momentum over the past few years. This review covers PARP imaging from the very first imaging agents up to the current state of the technology, with a focus on the clinical applications made possible by these agents.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Targeted PET imaging strategy to differentiate malignant from inflamed lymph nodes in diffuse large B-cell lymphoma

Jun Tang; Darin Salloum; Brandon Carney; Christian Brand; Susanne Kossatz; Ahmad Sadique; Jason S. Lewis; Wolfgang A. Weber; Hans-Guido Wendel; Thomas Reiner

Significance Diffuse large B-cell lymphoma (DLBCL) is the most common adult lymphoma, accounting for 37% of all non-Hodgkin lymphoma cases in the United States. Despite an approximate 50% cure rate, refractory or relapsed cases have a poor prognosis and require timely medical interventions. Therefore, accurate diagnostic methods play a pivotal role in managing DLBCL. 18F- fluorodeoxyglucose positron emission tomography ([18F]FDG-PET) imaging, the current standard imaging modality for diagnosing DLBCL, often fails to differentiate inflamed from malignant lymph nodes in patients with DLBCL. To address this urgent medical need, we have developed a targeted PET imaging method that accurately distinguishes malignancy from inflammation in the lymph nodes. Our targeted PET imaging approach could play an essential role in the clinical development of therapies that induce significant inflammation in DLBCL. Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma in adults. DLBCL exhibits highly aggressive and systemic progression into multiple tissues in patients, particularly in lymph nodes. Whole-body 18F-fluodeoxyglucose positron emission tomography ([18F]FDG-PET) imaging has an essential role in diagnosing DLBCL in the clinic; however, [18F]FDG-PET often faces difficulty in differentiating malignant tissues from certain nonmalignant tissues with high glucose uptake. We have developed a PET imaging strategy for DLBCL that targets poly[ADP ribose] polymerase 1 (PARP1), the expression of which has been found to be much higher in DLBCL than in healthy tissues. In a syngeneic DLBCL mouse model, this PARP1-targeted PET imaging approach allowed us to discriminate between malignant and inflamed lymph nodes, whereas [18F]FDG-PET failed to do so. Our PARP1-targeted PET imaging approach may be an attractive addition to the current PET imaging strategy to differentiate inflammation from malignancy in DLBCL.


Cancer Research | 2017

Biomarker based PET Imaging of Diffuse Intrinsic Pontine Glioma in Mouse Models.

Susanne Kossatz; Brandon Carney; Melanie Schweitzer; Giuseppe Carlucci; Vesselin Z. Miloushev; Uday Bhanu Maachani; Prajwal Rajappa; Kayvan R. Keshari; David Pisapia; Wolfgang A. Weber; Mark M. Souweidane; Thomas Reiner

Diffuse intrinsic pontine glioma (DIPG) is a childhood brainstem tumor with a universally poor prognosis. Here, we characterize a positron emission tomography (PET) probe for imaging DIPG in vivo In human histological tissues, the probes target, PARP1, was highly expressed in DIPG compared to normal brain. PET imaging allowed for the sensitive detection of DIPG in a genetically engineered mouse model, and probe uptake correlated to histologically determined tumor infiltration. Imaging with the sister fluorescence agent revealed that uptake was confined to proliferating, PARP1-expressing cells. Comparison with other imaging technologies revealed remarkable accuracy of our biomarker approach. We subsequently demonstrated that serial imaging of DIPG in mouse models enables monitoring of tumor growth, as shown in modeling of tumor progression. Overall, this validated method for quantifying DIPG burden would serve useful in monitoring treatment response in early phase clinical trials. Cancer Res; 77(8); 2112-23. ©2017 AACR.


PLOS ONE | 2016

Optical Imaging of PARP1 in Response to Radiation in Oral Squamous Cell Carcinoma.

Susanne Kossatz; Wolfgang A. Weber; Thomas Reiner

Targeting and inhibiting DNA repair pathways is a powerful strategy of controlling malignant growth. One such strategy includes the inhibition of PARP1, a central element in the intracellular DNA damage response. To determine and visualize the expression and intercellular distribution of PARP1 in vivo, and to monitor the pharmacokinetics of PARP1 targeted therapeutics, fluorescent small probes were developed. To date, however, it is unclear how these probes behave in a more realistic clinical setting, where DNA damage has been induced through one or more prior lines of therapy. Here, we use one such imaging agent, PARPi-FL, in tissues both with and without prior DNA damage, and investigate its value as a probe for PARP1 imaging. We show that PARP1 expression in oral cancer is high, and that the uptake of PARPi-FL is selective, irrespective of whether cells were exposed to irradiation or not. We also show that PARPi-FL uptake increases in response to DNA damage, and that this increase is reflected in higher enzyme expression. Our findings provide a framework for measuring exposure of cells to external beam radiation, and could help to elucidate the effects of such treatments non-invasively in mouse models of cancer.


ACS Medicinal Chemistry Letters | 2017

Synthesis of a Fluorescently Labeled 68Ga-DOTA-TOC Analog for Somatostatin Receptor Targeting

Sukhen C. Ghosh; Servando Hernandez Vargas; Melissa Rodriguez; Susanne Kossatz; Julie Voss; Kendra S. Carmon; Thomas Reiner; Agnes Schonbrunn; Ali Azhdarinia

Fluorescently labeled imaging agents can identify surgical margins in real-time to help achieve complete resections and minimize the likelihood of local recurrence. However, photon attenuation limits fluorescence-based imaging to superficial lesions or lesions that are a few millimeters beneath the tissue surface. Contrast agents that are dual-labeled with a radionuclide and fluorescent dye can overcome this limitation and combine quantitative, whole-body nuclear imaging with intraoperative fluorescence imaging. Using a multimodality chelation (MMC) scaffold, IRDye 800CW was conjugated to the clinically used somatostatin analog, 68Ga-DOTA-TOC, to produce the dual-labeled analog, 68Ga-MMC(IRDye 800CW)-TOC, with high yield and specific activity. In vitro pharmacological assays demonstrated retention of receptor-targeting properties for the dual-labeled compound with robust internalization that was somatostatin receptor (SSTR) 2-mediated. Biodistribution studies in mice identified the kidneys as the primary excretion route for 68Ga-MMC(IRDye 800CW)-TOC, along with clearance via the reticuloendothelial system. Higher uptake was observed in most tissues compared to 68Ga-DOTA-TOC but decreased as a function of time. The combination of excellent specificity for SSTR2-expressing cells and suitable biodistribution indicate potential application of 68Ga-MMC(IRDye 800CW)-TOC for intraoperative detection of SSTR2-expressing tumors.


The Journal of Nuclear Medicine | 2018

Direct Imaging of Drug Distribution and Target Engagement of the PARP Inhibitor Rucaparib

Susanne Kossatz; Brandon Carney; Christopher Farley; Wolfgang A. Weber; Charles Michael Drain; Thomas Reiner

Poly(ADP-ribose)polymerase (PARP) inhibitors have emerged as potent antitumor drugs. Here, we describe the intrinsic fluorescence properties of the clinically approved PARP inhibitor rucaparib and its potential to directly measure drug distribution and target engagement—a critical factor for understanding drug action and improving efficacy. Methods: We characterized the photophysical properties of rucaparib and determined its quantum yield and lifetime. Using confocal microscopy and flow cytometry, we imaged the intracellular distribution of rucaparib and measured uptake and release kinetics. Results: Rucaparib has an excitation/emission maximum of 355/480 nm and a quantum yield of 0.3. In vitro time-lapse imaging showed accumulation in cell nuclei within seconds of administration. Nuclear rucaparib uptake increased with higher PARP1 expression, and we determined an intracellular half-life of 6.4 h. Conclusion: The label-free, intrinsic fluorescence of rucaparib can be exploited to interrogate drug distribution and target binding, critical factors toward improving treatment efficacy and outcome.


Molecular Imaging | 2017

Detection and Delineation of Oral Cancer With a PARP1-Targeted Optical Imaging Agent

Susanne Kossatz; Wolfgang A. Weber; Thomas Reiner

More sensitive and specific methods for early detection are imperative to improve survival rates in oral cancer. However, oral cancer detection is still largely based on visual examination and histopathology of biopsy material, offering no molecular selectivity or spatial resolution. Intuitively, the addition of optical contrast could improve oral cancer detection and delineation, but so far no molecularly targeted approach has been translated. Our fluorescently labeled small-molecule inhibitor PARPi-FL binds to the DNA repair enzyme poly(ADP-ribose)polymerase 1 (PARP1) and is a potential diagnostic aid for oral cancer delineation. Based on our preclinical work, a clinical phase I/II trial opened in March 2017 to evaluate PARPi-FL as a contrast agent for oral cancer imaging. In this commentary, we discuss why we chose PARP1 as a biomarker for tumor detection and which particular characteristics make PARPi-FL an excellent candidate to image PARP1 in optically guided applications. We also comment on the potential benefits of our molecularly targeted PARPi-FL-guided imaging approach in comparison to existing oral cancer screening adjuncts and mention the adaptability of PARPi-FL imaging to other environments and tumor types.


The Journal of Nuclear Medicine | 2018

PARP-1–Targeted Radiotherapy in Mouse Models of Glioblastoma

Stephen A. Jannetti; Giuseppe Carlucci; Brandon Carney; Susanne Kossatz; Larissa Shenker; Lukas M. Carter; Beatriz Salinas; Christian Brand; Ahmad Sadique; Patrick L. Donabedian; Kristen M. Cunanan; Mithat Gonen; Vladimir Ponomarev; Brian M. Zeglis; Mark M. Souweidane; Jason S. Lewis; Wolfgang A. Weber; John L. Humm; Thomas Reiner

The DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP-1) is overexpressed in glioblastoma, with overall low expression in healthy brain tissue. Paired with the availability of specific small molecule inhibitors, PARP-1 is a near-ideal target to develop novel radiotherapeutics to induce DNA damage and apoptosis in cancer cells, while sparing healthy brain tissue. Methods: We synthesized an 131I-labeled PARP-1 therapeutic and investigated its pharmacology in vitro and in vivo. A subcutaneous tumor model was used to quantify retention times and therapeutic efficacy. A potential clinical scenario, intratumoral convection-enhanced delivery, was mimicked using an orthotopic glioblastoma model combined with an implanted osmotic pump system to study local administration of 131I-PARPi (PARPi is PARP inhibitor). Results: 131I-PARPi is a 1(2H)-phthalazinone, similar in structure to the Food and Drug Administration–approved PARP inhibitor AZD-2281. In vitro studies have shown that 131I-PARPi and AZD-2281 share similar pharmacologic profiles. 131I-PARPi delivered 134.1 cGy/MBq intratumoral injected activity. Doses to nontarget tissues, including liver and kidney, were significantly lower. Radiation damage and cell death in treated tumors were shown by p53 activation in U87-MG cells transfected with a p53-bioluminescent reporter. Treated mice showed significantly longer survival than mice receiving vehicle (29 vs. 22 d, P < 0.005) in a subcutaneous model. Convection-enhanced delivery demonstrated efficient retention of 131I-PARPi in orthotopic brain tumors, while quickly clearing from healthy brain tissue. Conclusion: Our results demonstrate 131I-PARPi’s high potential as a therapeutic and highlight PARP’s relevance as a target for radionuclide therapy. Radiation plays an integral role in brain tumor therapy, and radiolabeled PARP therapeutics could ultimately lead to improvements in the standard of care.


Archive | 2018

Molecular Imaging and Molecular Imaging Technologies

Katja Haedicke; Susanne Kossatz; Thomas Reiner; Jan Grimm

Molecular imaging has become an integral component of modern medicine. Defined by the molecular imaging center of excellence as “the visualization, characterization, and measurement of biological processes at the cellular and molecular level in humans and other living systems”, molecular imaging includes two- or three-dimensional noninvasive imaging as well as the quantification of acquired data over time [1]. Clinically, the importance of molecular imaging is paramount in the study and noninvasive diagnosis of diseases—based on molecular signatures rather than anatomic alterations—which in turn enables early detection of abnormalities and evaluation of new treatment approaches for improved survival rates. James and Gambhir summarized the main advantages of molecular imaging approaches: the possibility of studying cells in their natural environment without any disturbance from outside, the analysis of complex biological processes in real time, the investigation of signaling pathways in vivo, the gaining of information about drug delivery and pharmacokinetics, and the obtaining of multiple data sets in adequate resolution from the same patient over time [2].

Collaboration


Dive into the Susanne Kossatz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brandon Carney

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christian Brand

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Carlucci

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ahmad Sadique

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Beatriz Salinas

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher Irwin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mithat Gonen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Patrick L. Donabedian

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alexander Bolaender

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge