Network


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

Hotspot


Dive into the research topics where Jon G. Quatromoni is active.

Publication


Featured researches published by Jon G. Quatromoni.


Journal of Clinical Investigation | 2014

Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer

Evgeniy Eruslanov; Pratik Bhojnagarwala; Jon G. Quatromoni; Tom L. Stephen; Anjana Ranganathan; Charuhas Deshpande; Tatiana Akimova; Anil Vachani; Leslie A. Litzky; Wayne W. Hancock; Jose R. Conejo-Garcia; Michael Feldman; Steven M. Albelda; Sunil Singhal

Infiltrating inflammatory cells are highly prevalent within the tumor microenvironment and mediate many processes associated with tumor progression; however, the contribution of specific populations remains unclear. For example, the nature and function of tumor-associated neutrophils (TANs) in the cancer microenvironment is largely unknown. The goal of this study was to provide a phenotypic and functional characterization of TANs in surgically resected lung cancer patients. We found that TANs constituted 5%-25% of cells isolated from the digested human lung tumors. Compared with blood neutrophils, TANs displayed an activated phenotype (CD62L(lo)CD54(hi)) with a distinct repertoire of chemokine receptors that included CCR5, CCR7, CXCR3, and CXCR4. TANs produced substantial quantities of the proinflammatory factors MCP-1, IL-8, MIP-1α, and IL-6, as well as the antiinflammatory IL-1R antagonist. Functionally, both TANs and neutrophils isolated from distant nonmalignant lung tissue were able to stimulate T cell proliferation and IFN-γ release. Cross-talk between TANs and activated T cells led to substantial upregulation of CD54, CD86, OX40L, and 4-1BBL costimulatory molecules on the neutrophil surface, which bolstered T cell proliferation in a positive-feedback loop. Together our results demonstrate that in the earliest stages of lung cancer, TANs are not immunosuppressive, but rather stimulate T cell responses.


PLOS ONE | 2014

Intraoperative Near-Infrared Imaging Can Distinguish Cancer from Normal Tissue but Not Inflammation

Olugbenga T. Okusanya; Ryan Judy; Ollin Venegas; Jack Jiang; Elizabeth DeJesus; Evgeniy Eruslanov; Jon G. Quatromoni; Pratik Bhojnagarwala; Charuhas Deshpande; Steven M. Albelda; Shuming Nie; Sunil Singhal

Introduction Defining tumor from non-tumor tissue is one of the major challenges of cancer surgery. Surgeons depend on visual and tactile clues to select which tissues should be removed from a patient. Recently, we and others have hypothesized near-infrared (NIR) imaging can be used during surgery to differentiate tumors from normal tissue. Methods We enrolled 8 canines and 5 humans undergoing cancer surgery for NIR imaging. The patients were injected with indocyanine green (ICG), an FDA approved non-receptor specific NIR dye that accumulates in hyperpermeable tissues, 16–24 hours prior to surgery. During surgery, NIR imaging was used to discriminate the tumor from non-tumor tissue. Results NIR imaging identified all tumors with a mean signal-to-background ratio of 6.7. Optical images were useful during surgery in discriminating normal tissue from cancer. In 3 canine cases and 1 human case, the tissue surrounding the tumor was inflamed due to obstruction of the vascular supply due to mass effect. In these instances, NIR imaging could not distinguish tumor tissue from tissue that was congested, edematous and did not contain cancer. Conclusions This study shows that NIR imaging can identify tumors from normal tissues, provides excellent tissue contrast, and it facilitates the resection of tumors. However, in situations where there is significant peritumoral inflammation, NIR imaging with ICG is not helpful. This suggests that non-targeted NIR dyes that accumulate in hyperpermeable tissues will have significant limitations in the future, and receptor-specific NIR dyes may be necessary to overcome this problem.


OncoImmunology | 2013

Myeloid derived suppressor cells: Targets for therapy

Todd J. Waldron; Jon G. Quatromoni; Tatiana A. Karakasheva; Sunil Singhal; Anil K. Rustgi

The goal of achieving measurable response with cancer immunotherapy requires counteracting the immunosuppressive characteristics of tumors. One of the mechanisms that tumors utilize to escape immunosurveillance is the activation of myeloid derived suppressor cells (MDSCs). Upon activation by tumor-derived signals, MDSCs inhibit the ability of the host to mount an anti-tumor immune response via their capacity to suppress both the innate and adaptive immune systems. Despite their relatively recent discovery and characterization, anti-MDSC agents have been identified, which may improve immunotherapy efficacy.


Technology in Cancer Research & Treatment | 2015

Small portable interchangeable imager of fluorescence for fluorescence guided surgery and research.

Olugbenga T. Okusanya; Brian Madajewski; Erin Segal; Brendan F. Judy; Ollin Venegas; Ryan Judy; Jon G. Quatromoni; May D. Wang; Shuming Nie; Sunil Singhal

Fluorescence guided surgery (FGS) is a developing field of surgical and oncologic research. Practically, FGS has shown useful applications in urologic surgery, benign biliary surgery, colorectal cancer liver metastasis resection, and ovarian cancer debulking. Most notably in in cancer surgery, FGS allows for the clear delineation of cancerous tissue from benign tissue. FGS requires the utilization of a fluorescent contrast agent and an intraoperative fluorescence imaging device (IFID). Currently available IFIDs are expensive, unable to work with multiple fluorophores, and can be cumbersome. This study aims to describe the development and utility of a small, cost-efficient, and interchangeable IFID made from commercially available components. Extensive research was done to design and construct a light-weight, portable, and cost-effective IFID. We researched the capabilities, size, and cost of several camera types and eventually decided on a near-infrared (NIR) charged couple device (CCD) camera for its overall profile. The small portable interchangeable imager of fluorescence (SPIIF) is a “scout” IFID system for FGS. The main components of the SPIIF are a NIR CCD camera with an articulating light filter. These components and a LED light source with an attached heat sink are mounted on a small metal platform. The system is connected to a laptop by a USB 2.0 cable. Pixielink


Cancer Biology & Therapy | 2012

A positive-margin resection model recreates the postsurgical tumor microenvironment and is a reliable model for adjuvant therapy evaluation

Jarrod D. Predina; Brendan F. Judy; Zvi G. Fridlender; Louis A. Aliperti; Brian Madajewski; Veena Kapoor; Guanjun Cheng; Jon G. Quatromoni; Olugbenga T. Okusanya; Sunil Singhal

Up to 30% of cancer patients undergoing curative surgery develop local recurrences due to positive margins. Patients typically receive adjuvant chemotherapy, immunotherapy and/or radiation to prevent such relapses. Interestingly, evidence supporting these therapies is traditionally derived in animal models of primary tumors, thus failing to consider surgically induced tumor microenvironment changes that may influence adjuvant therapy efficacy. To address this consideration, we characterized a murine model of local cancer recurrence. This model was reproducible and generated a postoperative inflammatory tumor microenvironment that resembles those observed following human cancer surgery. To further validate this model, antagonists of two pro-inflammatory mediators, TGFβ and COX-2, were tested and found to be effective in decreasing the growth of recurrent tumors. We appreciated that preoperative TGFβ inhibition led to wound dehiscence, while postoperative initiation of COX-2 inhibition resulted in a loss of efficacy. In summary, although not an exact replica of all human cancer surgeries, our proposed local recurrence approach provides a biologically relevant and reliable model useful for preclinical evaluation of novel adjuvant therapies. The use of this model yields results that may be overlooked using traditional preclinical cancer models that fail to incorporate a surgical component.


Journal of Leukocyte Biology | 2015

An optimized disaggregation method for human lung tumors that preserves the phenotype and function of the immune cells

Jon G. Quatromoni; Sunil Singhal; Pratik Bhojnagarwala; Wayne W. Hancock; Steven M. Albelda; Evgeniy Eruslanov

Careful preparation of human tissues is the cornerstone of obtaining accurate data in immunologic studies. Despite the essential importance of tissue processing in tumor immunology and clinical medicine, current methods of tissue disaggregation have not been rigorously tested for data fidelity. Thus, we critically evaluated the current techniques available in the literature that are used to prepare human lung tumors for immunologic studies. We discovered that these approaches are successful at digesting cellular attachments and ECMs; however, these methods frequently alter the immune cell composition and/or expression of surface molecules. We thus developed a novel approach to prepare human lung tumors for immunologic studies by combining gentle mechanical manipulation with an optimized cocktail of enzymes used at low doses. This enzymatic digestion cocktail optimized cell yield and cell viability, retrieved all major tumor‐associated cell populations, and maintained the expression of cell‐surface markers for lineage definition and in vivo effector functions. To our knowledge, we present the first rigorously tested disaggregation method designed for human lung tumors.


Vascular and Endovascular Surgery | 2011

Impact of Intercostal Artery Occlusion on Spinal Cord Ischemia Following Thoracic Endovascular Aortic Repair

Brant W. Ullery; Jon G. Quatromoni; Benjamin M. Jackson; Edward Y. Woo; Ronald M. Fairman; Nimesh D. Desai; Joseph E. Bavaria; Grace J. Wang

Objective: To evaluate intercostal artery patency following thoracic endovascular aortic repair (TEVAR) and its relationship with spinal cord ischemia (SCI). Methods: Patients with SCI (n = 7) and a matched control cohort (n = 18) were identified from a prospectively maintained database. Radiographic analysis of intercostal patency was assessed using 3-dimensional (3-D)-reconstructed images of pre- and postoperative CT angiograms (1-6 months, 6-12 months, and 1-5 years). Results: Patients with SCI had a higher incidence of perioperative hypotension (P < .01) and longer procedure duration (P = .01). While the mean number of patent intercostal arteries at each time interval was not significantly different between groups, both SCI (P = .002) and control (P <.001) groups demonstrated a significant reduction in patent intercostal arteries in the stented area of the aorta following TEVAR. Conclusion: TEVAR decreases intercostal artery patency in the area of aortic coverage. Our data suggest that intercostal artery patency, in conjunction with perioperative hypotension, is an important contributor to postoperative SCI.


BMC Immunology | 2013

The timing of TGF-β inhibition affects the generation of antigen-specific CD8+ T Cells

Jon G. Quatromoni; Eiji Suzuki; Olugbenga T. Okusanya; Brendan F. Judy; Pratik Bhojnagarwala; Ollin Venegas; Evgeniy Eruslanov; Jarrod D. Predina; Steven M. Albelda; Sunil Singhal

BackgroundTransforming growth factor (TGF)-β is a potent immunosuppressive cytokine necessary for cancer growth. Animal and human studies have shown that pharmacologic inhibition of TGF-β slows the growth rate of established tumors and occasionally eradicates them altogether. We observed, paradoxically, that inhibiting TGF-β before exposing animals to tumor cells increases tumor growth kinetics. We hypothesized that TGF-β is necessary for the anti-tumor effects of cytotoxic CD8+ T lymphocytes (CTLs) during the early stages of tumor initiation.MethodsBALB/c mice were pretreated with a blocking soluble TGF-β receptor (sTGF-βR, TGF-β-blockade group, n=20) or IgG2a (Control group, n=20) before tumor inoculation. Tumor size was followed for 6 weeks. In vivo lymphocyte assays and depletion experiments were then performed to investigate the immunological basis of our results. Lastly, animals were pretreated with either sTGF-βR (n=6) or IgG2a (n=6) prior to immunization with an adenoviral vector encoding the human papillomavirus E7 gene (Ad.E7). One week later, flow cytometry was utilized to measure the number of splenic E7-specific CD8+ T cells.ResultsInhibition of TGF-β before the injection of tumor cells resulted in significantly larger average tumor volumes on days 11, 17, 22, 26 and 32 post tumor-inoculation (p < 0.05). This effect was due to the inhibition of CTLs, as it was not present in mice with severe combined immunodeficiency (SCID) or those depleted of CD8+ T cells. Furthermore, pretreatment with sTGF-βR inhibited tumor-specific CTL activity in a Winn Assay. Tumors grew to a much larger size when mixed with CD8+ T cells from mice pretreated with sTGF-βR than when mixed with CD8+ T cells from mice in the control group: 96 mm3 vs. 22.5 mm3, respectively (p < 0.05). In addition, fewer CD8+ T cells were generated in Ad.E7-immunized mice pretreated with sTGF-βR than in mice from the control group: 0.6% total CD8+ T cells vs. 1.9%, respectively (p < 0.05).ConclusionsThese studies provide the first in vivo evidence that TGF-β may be necessary for anti-tumor immune responses in certain cancers. This finding has important implications for our understanding of anti-tumor immune responses, the role of TGF-β in the immune system, and the future development of TGF-β inhibiting drugs.


Seminars in Interventional Radiology | 2015

Advanced Endovascular Approaches in the Management of Challenging Proximal Aortic Neck Anatomy: Traditional Endografts and the Snorkel Technique.

Jon G. Quatromoni; Ksenia A. Orlova; Paul J. Foley

Advances in endovascular technology, and access to this technology, have significantly changed the field of vascular surgery. Nowhere is this more apparent than in the treatment of abdominal aortic aneurysms (AAAs), in which endovascular aneurysm repair (EVAR) has replaced the traditional open surgical approach in patients with suitable anatomy. However, approximately one-third of patients presenting with AAAs are deemed ineligible for standard EVAR because of anatomic constraints, the majority of which involve the proximal aneurysmal neck. To overcome these challenges, a bevy of endovascular approaches have been developed to either enhance stent graft fixation at the proximal neck or extend the proximal landing zone to allow adequate apposition to the aortic wall and thus aneurysm exclusion. This article is composed of two sections that together address new endovascular approaches for treating aortic aneurysms with difficult proximal neck anatomy. The first section will explore advancements in the traditional EVAR approach for hostile neck anatomy that maximize the use of the native proximal landing zone; the second section will discuss a technique that was developed to extend the native proximal landing zone and maintain perfusion to vital aortic branches using common, off-the-shelf components: the snorkel technique. While the techniques presented differ in terms of approach, the available clinical data, albeit limited, support the notion that they may both have roles in the treatment algorithm for patients with challenging proximal neck anatomy.


Journal of Immunotherapy | 2014

Adenoviral-based immunotherapy provides local disease control in an orthotopic murine model of esophageal cancer.

Jon G. Quatromoni; Jarrod D. Predina; Pratik Bhojnagarwala; Ryan Judy; Jack Jiang; Elizabeth M. De Jesus; Veena Kapoor; Guanjun Cheng; Olugbenga T. Okusanya; Evgeniy Eruslanov; Sunil Singhal

Despite recent advances in the development of novel therapies, esophageal carcinoma remains an aggressive cancer associated with a poor prognosis. The lack of a high throughput, reproducible syngeneic animal model that replicates human disease is partly responsible for the paucity of novel therapeutic approaches. In this report, we present the first successful syngeneic, orthotopic model for esophageal cancer. This model was used to test an established adenoviral-based tumor vaccine. We utilized a murine esophageal cancer cell line established from the ED-L2-cyclin D1;p53–/– mouse that was transduced to express a viral tumor antigen, the Human Papilloma Virus (HPV) E7 protein. The tumor was established in its natural microenvironment at the gastroesophageal junction. Tumor growth was consistent and reproducible. An adenoviral vaccine to E7 (Ad.E7) induced an E7-specific population of functionally active CD8+ T cells that trafficked into the tumors and retained cytotoxicity. Ad.E7 vaccination reduced local tumor growth and prolonged overall survival. These findings suggest that orthotopic tumor growth is a reasonable preclinical model to validate novel therapies.

Collaboration


Dive into the Jon G. Quatromoni's collaboration.

Top Co-Authors

Avatar

Sunil Singhal

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Evgeniy Eruslanov

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven M. Albelda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wayne W. Hancock

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Feldman

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Tatiana Akimova

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge