Network


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

Hotspot


Dive into the research topics where David Calligaris is active.

Publication


Featured researches published by David Calligaris.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Intraoperative mass spectrometry mapping of an onco-metabolite to guide brain tumor surgery

Sandro Santagata; Livia S. Eberlin; Isaiah Norton; David Calligaris; Daniel R. Feldman; Jennifer L. Ide; Xiaohui Liu; Joshua S. Wiley; Matthew L. Vestal; Shakti Ramkissoon; Daniel A. Orringer; Kristen K. Gill; Ian F. Dunn; Dora Dias-Santagata; Keith L. Ligon; Ferenc A. Jolesz; Alexandra J. Golby; R. Graham Cooks; Nathalie Y. R. Agar

Significance The diagnosis of tumors during surgery still relies principally on an approach developed over 150 y ago: frozen section microscopy. We show that a validated molecular marker—2-hydroxyglutarate generated from isocitrate dehydrogenase 1 mutant gliomas—can be rapidly detected from tumors using a form of ambient MS that does not require sample preparation. We use the Advanced Multimodality Image Guided Operating Suite at Brigham and Women’s Hospital to demonstrate that desorption electrospray ionization MS could be used to detect residual tumor that would have been left behind in the patient. The approach paves the way for the clinical testing of MS-based intraoperative monitoring of tumor metabolites, an advance that could revolutionize the care of surgical oncology patients. For many intraoperative decisions surgeons depend on frozen section pathology, a technique developed over 150 y ago. Technical innovations that permit rapid molecular characterization of tissue samples at the time of surgery are needed. Here, using desorption electrospray ionization (DESI) MS, we rapidly detect the tumor metabolite 2-hydroxyglutarate (2-HG) from tissue sections of surgically resected gliomas, under ambient conditions and without complex or time-consuming preparation. With DESI MS, we identify isocitrate dehydrogenase 1-mutant tumors with both high sensitivity and specificity within minutes, immediately providing critical diagnostic, prognostic, and predictive information. Imaging tissue sections with DESI MS shows that the 2-HG signal overlaps with areas of tumor and that 2-HG levels correlate with tumor content, thereby indicating tumor margins. Mapping the 2-HG signal onto 3D MRI reconstructions of tumors allows the integration of molecular and radiologic information for enhanced clinical decision making. We also validate the methodology and its deployment in the operating room: We have installed a mass spectrometer in our Advanced Multimodality Image Guided Operating (AMIGO) suite and demonstrate the molecular analysis of surgical tissue during brain surgery. This work indicates that metabolite-imaging MS could transform many aspects of surgical care.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Application of desorption electrospray ionization mass spectrometry imaging in breast cancer margin analysis

David Calligaris; Diana Caragacianu; Xiaohui Liu; Isaiah Norton; Christopher J. Thompson; Andrea L. Richardson; Mehra Golshan; Michael L. Easterling; Sandro Santagata; Deborah A. Dillon; Ferenc A. Jolesz; Nathalie Y. R. Agar

Significance This study is the first demonstration, to our knowledge, of the application of desorption electrospray ionization mass spectrometry imaging (DESI-MSI) for discrimination of breast cancer and delineation of tumor margins. Using DESI-MSI, it is possible to discriminate between cancerous and adjacent normal tissue on the basis of the detection and specific spatial distributions of different lipid species. This study proves the feasibility of classifying cancerous and normal breast tissues using ambient ionization MSI. It will allow the surgeon to access to this information in real time so as to make accurate intraoperative decisions quickly. It will result in improved cosmesis and decrease the need for multiple operations for margin reexcision. Distinguishing tumor from normal glandular breast tissue is an important step in breast-conserving surgery. Because this distinction can be challenging in the operative setting, up to 40% of patients require an additional operation when traditional approaches are used. Here, we present a proof-of-concept study to determine the feasibility of using desorption electrospray ionization mass spectrometry imaging (DESI-MSI) for identifying and differentiating tumor from normal breast tissue. We show that tumor margins can be identified using the spatial distributions and varying intensities of different lipids. Several fatty acids, including oleic acid, were more abundant in the cancerous tissue than in normal tissues. The cancer margins delineated by the molecular images from DESI-MSI were consistent with those margins obtained from histological staining. Our findings prove the feasibility of classifying cancerous and normal breast tissues using ambient ionization MSI. The results suggest that an MS-based method could be developed for the rapid intraoperative detection of residual cancer tissue during breast-conserving surgery.


Clinical Cancer Research | 2015

The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood–Brain Barrier in Glioblastoma

Jenny L. Pokorny; David Calligaris; Shiv K. Gupta; Dennis O. Iyekegbe; Dustin Mueller; Katrina Bakken; Brett L. Carlson; Mark A. Schroeder; Debra Evans; Zhenkun Lou; Paul A. Decker; Jeanette E. Eckel-Passow; Vincenzo Pucci; Bennett Ma; Stuart D. Shumway; William F. Elmquist; Nathalie Y. R. Agar; Jann N. Sarkaria

Purpose: Wee1 regulates key DNA damage checkpoints, and in this study, the efficacy of the Wee1 inhibitor MK-1775 was evaluated in glioblastoma multiforme (GBM) xenograft models alone and in combination with radiation and/or temozolomide. Experimental Design: In vitro MK-1775 efficacy alone and in combination with temozolomide, and the impact on DNA damage, was analyzed by Western blotting and γH2AX foci formation. In vivo efficacy was evaluated in orthotopic and heterotopic xenografts. Drug distribution was assessed by conventional mass spectrometry (MS) and matrix-assisted laser desorption/ionization (MALDI)-MS imaging. Results: GBM22 (IC50 = 68 nmol/L) was significantly more sensitive to MK-1775 compared with five other GBM xenograft lines, including GBM6 (IC50 >300 nmol/L), and this was associated with a significant difference in pan-nuclear γH2AX staining between treated GBM22 (81% cells positive) and GBM6 (20% cells positive) cells. However, there was no sensitizing effect of MK-1775 when combined with temozolomide in vitro. In an orthotopic GBM22 model, MK-1775 was ineffective when combined with temozolomide, whereas in a flank model of GBM22, MK-1775 exhibited both single-agent and combinatorial activity with temozolomide. Consistent with limited drug delivery into orthotopic tumors, the normal brain to whole blood ratio following a single MK-1775 dose was 5%, and MALDI-MS imaging demonstrated heterogeneous and markedly lower MK-1775 distribution in orthotopic as compared with heterotopic GBM22 tumors. Conclusions: Limited distribution to brain tumors may limit the efficacy of MK-1775 in GBM. Clin Cancer Res; 21(8); 1916–24. ©2015 AACR.


Journal of Mass Spectrometry | 2013

Mass Spectrometry Imaging as a Tool for Surgical Decision-Making

David Calligaris; Isaiah Norton; Daniel R. Feldman; Jennifer L. Ide; Ian F. Dunn; Livia S. Eberlin; R. G. Cooks; Ferenc A. Jolesz; Alexandra J. Golby; Sandro Santagata; Nathalie Y. R. Agar

Despite significant advances in image-guided therapy, surgeons are still too often left with uncertainty when deciding to remove tissue. This binary decision between removing and leaving tissue during surgery implies that the surgeon should be able to distinguish tumor from healthy tissue. In neurosurgery, current image-guidance approaches such as magnetic resonance imaging (MRI) combined with neuronavigation offer a map as to where the tumor should be, but the only definitive method to characterize the tissue at stake is histopathology. Although extremely valuable information is derived from this gold standard approach, it is limited to very few samples during surgery and is not practically used for the delineation of tumor margins. The development and implementation of faster, comprehensive, and complementary approaches for tissue characterization are required to support surgical decision-making--an incremental and iterative process with tumor removed in multiple and often minute biopsies. The development of atmospheric pressure ionization sources makes it possible to analyze tissue specimens with little to no sample preparation. Here, we highlight the value of desorption electrospray ionization as one of many available approaches for the analysis of surgical tissue. Twelve surgical samples resected from a patient during surgery were analyzed and diagnosed as glioblastoma tumor or necrotic tissue by standard histopathology, and mass spectrometry results were further correlated to histopathology for critical validation of the approach. The use of a robust statistical approach reiterated results from the qualitative detection of potential biomarkers of these tissue types. The correlation of the mass spectrometry and histopathology results to MRI brings significant insight into tumor presentation that could not only serve to guide tumor resection, but that is also worthy of more detailed studies on our understanding of tumor presentation on MRI.


Cancer Research | 2016

Label-Free Neurosurgical Pathology with Stimulated Raman Imaging

Fake Lu; David Calligaris; Olutayo Olubiyi; Isaiah Norton; Wenlong Yang; Sandro Santagata; X. Sunney Xie; Alexandra J. Golby; Nathalie Y. R. Agar

The goal of brain tumor surgery is to maximize tumor removal without injuring critical brain structures. Achieving this goal is challenging as it can be difficult to distinguish tumor from nontumor tissue. While standard histopathology provides information that could assist tumor delineation, it cannot be performed iteratively during surgery as freezing, sectioning, and staining of the tissue require too much time. Stimulated Raman scattering (SRS) microscopy is a powerful label-free chemical imaging technology that enables rapid mapping of lipids and proteins within a fresh specimen. This information can be rendered into pathology-like images. Although this approach has been used to assess the density of glioma cells in murine orthotopic xenografts models and human brain tumors, tissue heterogeneity in clinical brain tumors has not yet been fully evaluated with SRS imaging. Here we profile 41 specimens resected from 12 patients with a range of brain tumors. By evaluating large-scale stimulated Raman imaging data and correlating this data with current clinical gold standard of histopathology for 4,422 fields of view, we capture many essential diagnostic hallmarks for glioma classification. Notably, in fresh tumor samples, we observe additional features, not seen by conventional methods, including extensive lipid droplets within glioma cells, collagen deposition in gliosarcoma, and irregularity and disruption of myelinated fibers in areas infiltrated by oligodendroglioma cells. The data are freely available in a public resource to foster diagnostic training and to permit additional interrogation. Our work establishes the methodology and provides a significant collection of reference images for label-free neurosurgical pathology. Cancer Res; 76(12); 3451-62. ©2016 AACR.


Molecular Cancer Therapeutics | 2015

Efficacy of PARP Inhibitor Rucaparib in Orthotopic Glioblastoma Xenografts Is Limited by Ineffective Drug Penetration into the Central Nervous System

Karen E. Parrish; Ling Cen; James Murray; David Calligaris; Sani H. Kizilbash; Rajendar K. Mittapalli; Brett L. Carlson; Mark A. Schroeder; Julieann Sludden; Alan V. Boddy; Nathalie Y. R. Agar; Nicola J. Curtin; William F. Elmquist; Jann N. Sarkaria

PARP inhibition can enhance the efficacy of temozolomide and prolong survival in orthotopic glioblastoma (GBM) xenografts. The aim of this study was to evaluate the combination of the PARP inhibitor rucaparib with temozolomide and to correlate pharmacokinetic and pharmacodynamic studies with efficacy in patient-derived GBM xenograft models. The combination of rucaparib with temozolomide was highly effective in vitro in short-term explant cultures derived from GBM12, and, similarly, the combination of rucaparib and temozolomide (dosed for 5 days every 28 days for 3 cycles) significantly prolonged the time to tumor regrowth by 40% in heterotopic xenografts. In contrast, the addition of rucaparib had no impact on the efficacy of temozolomide in GBM12 or GBM39 orthotopic models. Using Madin-Darby canine kidney (MDCK) II cells stably expressing murine BCRP1 or human MDR1, cell accumulation studies demonstrated that rucaparib is transported by both transporters. Consistent with the influence of these efflux pumps on central nervous system drug distribution, Mdr1a/b−/−Bcrp1−/− knockout mice had a significantly higher brain to plasma ratio for rucaparib (1.61 ± 0.25) than wild-type mice (0.11 ± 0.08). A pharmacokinetic and pharmacodynamic evaluation after a single dose confirmed limited accumulation of rucaparib in the brain is associated with substantial residual PARP enzymatic activity. Similarly, matrix-assisted laser desorption/ionization mass spectrometric imaging demonstrated significantly enhanced accumulation of drug in flank tumor compared with normal brain or orthotopic tumors. Collectively, these results suggest that limited drug delivery into brain tumors may significantly limit the efficacy of rucaparib combined with temozolomide in GBM. Mol Cancer Ther; 14(12); 2735–43. ©2015 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

MALDI mass spectrometry imaging analysis of pituitary adenomas for near-real-time tumor delineation

David Calligaris; Daniel R. Feldman; Isaiah Norton; Olutayo Olubiyi; Armen Changelian; Revaz Machaidze; Matthew L. Vestal; Edward R. Laws; Ian F. Dunn; Sandro Santagata; Nathalie Y. R. Agar

Significance This study presents the use of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) to detect and delineate pituitary tumors. Using MALDI MSI, it is possible to determine the peptide and protein hormone composition of pituitary tumor resection samples in fewer than 30 min. Surgeons could therefore have access to critical information for surgical decision-making in a near-real-time manner and be able to localize and discriminate pituitary tumor from nonpathological pituitary gland. This study supports the inclusion of MALDI MSI in the clinical workflow for the surgical resection of pituitary tumors, potentially allowing for improved surgical precision and patient outcomes. We present a proof of concept study designed to support the clinical development of mass spectrometry imaging (MSI) for the detection of pituitary tumors during surgery. We analyzed by matrix-assisted laser desorption/ionization (MALDI) MSI six nonpathological (NP) human pituitary glands and 45 hormone secreting and nonsecreting (NS) human pituitary adenomas. We show that the distribution of pituitary hormones such as prolactin (PRL), growth hormone (GH), adrenocorticotropic hormone (ACTH), and thyroid stimulating hormone (TSH) in both normal and tumor tissues can be assessed by using this approach. The presence of most of the pituitary hormones was confirmed by using MS/MS and pseudo-MS/MS methods, and subtyping of pituitary adenomas was performed by using principal component analysis (PCA) and support vector machine (SVM). Our proof of concept study demonstrates that MALDI MSI could be used to directly detect excessive hormonal production from functional pituitary adenomas and generally classify pituitary adenomas by using statistical and machine learning analyses. The tissue characterization can be completed in fewer than 30 min and could therefore be applied for the near-real-time detection and delineation of pituitary tumors for intraoperative surgical decision-making.


Nature Communications | 2017

Blood-brain-barrier spheroids as an in vitro screening platform for brain-penetrating agents

Choi-Fong Cho; Justin Wolfe; Colin M. Fadzen; David Calligaris; Kalvis Hornburg; E. Antonio Chiocca; Nathalie Y. R. Agar; Bradley L. Pentelute; Sean E. Lawler

Culture-based blood–brain barrier (BBB) models are crucial tools to enable rapid screening of brain-penetrating drugs. However, reproducibility of in vitro barrier properties and permeability remain as major challenges. Here, we report that self-assembling multicellular BBB spheroids display reproducible BBB features and functions. The spheroid core is comprised mainly of astrocytes, while brain endothelial cells and pericytes encase the surface, acting as a barrier that regulates transport of molecules. The spheroid surface exhibits high expression of tight junction proteins, VEGF-dependent permeability, efflux pump activity and receptor-mediated transcytosis of angiopep-2. In contrast, the transwell co-culture system displays comparatively low levels of BBB regulatory proteins, and is unable to discriminate between the transport of angiopep-2 and a control peptide. Finally, we have utilized the BBB spheroids to screen and identify BBB-penetrant cell-penetrating peptides (CPPs). This robust in vitro BBB model could serve as a valuable next-generation platform for expediting the development of CNS therapeutics.


Analytical and Bioanalytical Chemistry | 2015

Profiling of adrenocorticotropic hormone and arginine vasopressin in human pituitary gland and tumor thin tissue sections using droplet-based liquid-microjunction surface-sampling-HPLC–ESI-MS–MS

Vilmos Kertesz; David Calligaris; Daniel R. Feldman; Armen Changelian; Edward R. Laws; Sandro Santagata; Nathalie Y. R. Agar; Gary J. Van Berkel

AbstractDescribed here are the results from the profiling of the proteins arginine vasopressin (AVP) and adrenocorticotropic hormone (ACTH) from normal human pituitary gland and pituitary adenoma tissue sections, using a fully automated droplet-based liquid-microjunction surface-sampling-HPLC–ESI-MS–MS system for spatially resolved sampling, HPLC separation, and mass spectrometric detection. Excellent correlation was found between the protein distribution data obtained with this method and data obtained with matrix-assisted laser desorption/ionization (MALDI) chemical imaging analyses of serial sections of the same tissue. The protein distributions correlated with the visible anatomic pattern of the pituitary gland. AVP was most abundant in the posterior pituitary gland region (neurohypophysis), and ATCH was dominant in the anterior pituitary gland region (adenohypophysis). The relative amounts of AVP and ACTH sampled from a series of ACTH-secreting and non-secreting pituitary adenomas correlated with histopathological evaluation. ACTH was readily detected at significantly higher levels in regions of ACTH-secreting adenomas and in normal anterior adenohypophysis compared with non-secreting adenoma and neurohypophysis. AVP was mostly detected in normal neurohypophysis, as expected. This work reveals that a fully automated droplet-based liquid-microjunction surface-sampling system coupled to HPLC–ESI-MS–MS can be readily used for spatially resolved sampling, separation, detection, and semi-quantitation of physiologically-relevant peptide and protein hormones, including AVP and ACTH, directly from human tissue. In addition, the relative simplicity, rapidity, and specificity of this method support the potential of this basic technology, with further advancement, for assisting surgical decision-making. Graphical AbstractMass spectrometry based profiling of hormones in human pituitary gland and tumor thin tissue sections


Oncotarget | 2017

Multiple spatially related pharmacophores define small molecule inhibitors of OLIG2 in glioblastoma.

Igor Tsigelny; Rajesh Mukthavaram; Valentina L. Kouznetsova; Ying Chao; Ivan Babic; Elmar Nurmemmedov; Sandra Pastorino; Pengfei Jiang; David Calligaris; Nathalie Y. R. Agar; Miriam Scadeng; Sandeep C. Pingle; Wolfgang Wrasidlo; Milan Makale; Santosh Kesari

Transcription factors (TFs) are a major class of protein signaling molecules that play key cellular roles in cancers such as the highly lethal brain cancer—glioblastoma (GBM). However, the development of specific TF inhibitors has proved difficult owing to expansive protein-protein interfaces and the absence of hydrophobic pockets. We uniquely defined the dimerization surface as an expansive parental pharmacophore comprised of several regional daughter pharmacophores. We targeted the OLIG2 TF which is essential for GBM survival and growth, we hypothesized that small molecules able to fit each subpharmacophore would inhibit OLIG2 activation. The most active compound was OLIG2 selective, it entered the brain, and it exhibited potent anti-GBM activity in cell-based assays and in pre-clinical mouse orthotopic models. These data suggest that (1) our multiple pharmacophore approach warrants further investigation, and (2) our most potent compounds merit detailed pharmacodynamic, biophysical, and mechanistic characterization for potential preclinical development as GBM therapeutics.

Collaboration


Dive into the David Calligaris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandro Santagata

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Isaiah Norton

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Daniel R. Feldman

Lawrence Berkeley National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Ian F. Dunn

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexandra J. Golby

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ferenc A. Jolesz

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge