Network


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

Hotspot


Dive into the research topics where Brendan F. Judy is active.

Publication


Featured researches published by Brendan F. Judy.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Changes in the local tumor microenvironment in recurrent cancers may explain the failure of vaccines after surgery

Jarrod D. Predina; Evgeniy Eruslanov; Brendan F. Judy; Veena Kapoor; Guanjun Cheng; Liang-Chuan Wang; Jing Sun; Edmund Moon; Zvi G. Fridlender; Steven M. Albelda; Sunil Singhal

Each year, more than 700,000 people undergo cancer surgery in the United States. However, more than 40% of those patients develop recurrences and have a poor outcome. Traditionally, the medical community has assumed that recurrent tumors arise from selected tumor clones that are refractory to therapy. However, we found that tumor cells have few phenotypical differences after surgery. Thus, we propose an alternative explanation for the resistance of recurrent tumors. Surgery promotes inhibitory factors that allow lingering immunosuppressive cells to repopulate small pockets of residual disease quickly. Recurrent tumors and draining lymph nodes are infiltrated with M2 (CD11b+F4/80hiCD206hi and CD11b+F4/80hiCD124hi) macrophages and CD4+Foxp3+ regulatory T cells. This complex network of immunosuppression in the surrounding tumor microenvironment explains the resistance of tumor recurrences to conventional cancer vaccines despite small tumor size, an intact antitumor immune response, and unaltered cancer cells. Therapeutic strategies coupling antitumor agents with inhibition of immunosuppressive cells potentially could impact the outcomes of more than 250,000 people each year.


Clinical Cancer Research | 2012

Intraoperative Near-Infrared Imaging of Surgical Wounds after Tumor Resections Can Detect Residual Disease

Brian Madajewski; Brendan F. Judy; Anas Mouchli; Veena Kapoor; May D. Wang; Shuming Nie; Sunil Singhal

Purpose: Surgical resection remains the most effective therapy for solid tumors worldwide. The most important prognostic indicator for cure following cancer surgery is a complete resection with no residual disease. However, intraoperative detection of retained cancer cells after surgery is challenging, and residual disease continues to be the most common cause of local failure. We hypothesized that visual enhancement of tumors using near-infrared imaging could potentially identify tumor deposits in the wound after resection. Experimental Design: A small animal model of surgery and retained disease was developed. Residual tumor deposits in the wound were targeted using an U.S. Food and Drug Administration–approved imaging agent, indocyanine green, by the enhanced permeability and retention effect. A novel handheld spectrometer was used to optically visualize retained disease after surgery. Results: We found residual disease using near-infrared imaging during surgery that was not visible to the naked eye or micro-CT. Furthermore, examination of tumor nodules was remarkably precise in delineating margins from normal surrounding tissues. This approach was most successful for tumors with increased neovasculature. Conclusions: The results suggest that near-infrared examination of the surgical wound after curative resection can potentially enable the surgeon to locate residual disease. The data in this study is the basis of an ongoing Phase I/II clinical trial in patients who undergo resection for lung and breast cancer. Clin Cancer Res; 18(20); 5741–51. ©2012 AACR.


Journal of Hematology & Oncology | 2012

Cytoreduction surgery reduces systemic myeloid suppressor cell populations and restores intratumoral immunotherapy effectiveness.

Jarrod D. Predina; Veena Kapoor; Brendan F. Judy; Guanjun Cheng; Zvi G. Fridlender; Steven M. Albelda; Sunil Singhal

BackgroundMultiple immunotherapy approaches have improved adaptive anti-tumor immune responses in patients with early stage disease; however, results have been less dramatic when treating patients with late stage disease. These blunted responses are likely due to a host of factors, including changes in the tumor microenvironment and systemic immunosuppressive features, which accompany advanced tumor states. We hypothesized that cytoreductive surgery could control these immunosuppressive networks and restore the potency of immunotherapy in advanced disease scenarios.MethodsTo test these hypotheses, two representative intratumoral immunotherapies (an adenoviral vector encoding a suicide gene, AdV-tk, or a type-I interferon, Ad.IFNα) were tested in murine models of lung cancer. Cytoreductive surgery was performed following treatment of advanced tumors. Mechanistic underpinnings were investigated using flow cytometry, in vivo leukocyte depletion methods and in vivo tumor neutralization assays.ResultsAdV-tk and Ad.IFNα were effective in treating early lung cancers, but had little anti-tumor effects in late stage cancers. Interestingly, in late stage scenarios, surgical cytoreduction unmasked the anti-tumor potency of both immunotherapeutic approaches. Immune mechanisms that explained restoration in anti-tumor immune responses included increased CD8 T-cell trafficking and reduced myeloid derived suppressor cell populations.ConclusionThis study demonstrates that surgical resection combined with immunotherapy may be a rational therapeutic option for patients with advanced stage cancer.


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 Gene Therapy | 2011

Neoadjuvant in situ gene-mediated cytotoxic immunotherapy improves postoperative outcomes in novel syngeneic esophageal carcinoma models

Jarrod D. Predina; Brendan F. Judy; Louis A. Aliperti; Zvi G. Fridlender; Aaron Blouin; Veena Kapoor; Benjamin A. Laguna; H Nakagawa; Anil K. Rustgi; Laura K. Aguilar; Estuardo Aguilar-Cordova; Steven M. Albelda; Sunil Singhal

Esophageal carcinoma is the most rapidly increasing tumor in the United States and has a dismal 15% 5-year survival. Immunotherapy has been proposed to improve patient outcomes; however, no immunocompetent esophageal carcinoma model exists to date to test this approach. We developed two mouse models of esophageal cancer by inoculating immunocompetent mice with syngeneic esophageal cell lines transformed by cyclin-D1 or mutant HRASG12V and loss of p53. Similar to humans, surgery and adjuvant chemotherapy (cisplatin and 5-fluorouracil) demonstrated limited efficacy. Gene-mediated cyototoxic immunotherapy (adenoviral vector carrying the herpes simplex virus thymidine kinase gene in combination with the prodrug ganciclovir; AdV-tk/GCV) demonstrated high levels of in vitro transduction and efficacy. Using in vivo syngeneic esophageal carcinoma models, combining surgery, chemotherapy and AdV-tk/GCV improved survival (P=0.007) and decreased disease recurrence (P<0.001). Mechanistic studies suggested that AdV-tk/GCV mediated a direct cytotoxic effect and an increased intra-tumoral trafficking of CD8 T cells (8.15% vs 14.89%, P=0.02). These data provide the first preclinical evidence that augmenting standard of care with immunotherapy may improve outcomes in the management of esophageal carcinoma.


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.


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.


Immunotherapy | 2012

How can cytoreduction surgery improve the prospects for cancer patients receiving immunotherapy

Brendan F. Judy; Sunil Singhal

Cancer, the leading cause of death for people under the age of 85 years in the USA, claims over 1500 lives per day [1,2]. The four most common cancers (lung, colorectal, prostate and breast cancer) account for more than half of total cancer incidences in the USA [2]. The most common cancer therapies are surgery, chemotherapy and radiation [2]. Except for a few chemotherapeutic regimens, surgery is the only definitive treatment available. Unfortunately, surgery has a 40–60% recurrence rate depending on the type of cancer [2]. This high recurrence rate has led to the combination of surgery with traditional therapies such as chemotherapy, immunotherapy and radiation. Immunotherapy utilizes the body’s own immune system to target cancer cells. Almost a century ago, it was hypothesized that a tumor can be recognized as foreign by the host and thus the immune system can be effective in eliminating these foreign cells [3]. Since then, the cancer immune response has been confirmed in knockout mice [4] and the exact phases of this mechanism, termed immunoediting, are still being rigorously studied [5]. Immunotherapy is advantageous because of its low toxicity, tumor specificity and ability to potentially produce long-term immunity. Immunotherapy seeks to eliminate cancer through both active and passive approaches [6]. Active immunotherapy utilizes our knowledge of tumor antigen expression and exploits endogenous, immunological processes of T-cell induction to eliminate the tumor [7]. Regardless of the modality, the primary goal of active immunotherapy is to prime and induce antigen-specific T-cell responses in order to eradicate residual disease and prevent postoperative recurrences. On the other hand, passive immunotherapy involves the use of immunologically active agents that are capable of exerting anti-tumor effects independently from the host’s immune system [7]. Such passive approaches include adoptive T-cell transfer techniques, which involve ex vivo amplification and reintroduction of autologous cytokine-induced killer or lymphokine-activated killer cells [7]. Various mechanisms by which immunotherapy reduces tumor burden have been elucidated, including increased intratumoral neutrophils, enhanced CD8 T-cell trafficking and decreased vasculature of the tumor [8,9].


Immunology Letters | 2015

Surgical cytoreduction restores the antitumor efficacy of a Listeria monocytogenes vaccine in malignant pleural mesothelioma

Gregory T. Kennedy; Brendan F. Judy; Pratik Bhojnagarwala; Edmund Moon; Zvi G. Fridlender; Steven M. Albelda; Sunil Singhal

Recent studies suggest that immunotherapy may offer a promising treatment strategy for early-stage malignant pleural mesothelioma (MPM), but advanced tumor burden may limit the efficacy of immunotherapy. Therefore, we hypothesized that surgical cytoreduction could restore the efficacy of vaccine-based immunotherapy for MPM. We developed a murine model of MPM through transduction of a mesothelioma cell line with mesothelin. We used this model to evaluate the efficacy of a Listeria monocytogenes vaccine expressing mesothelin. Tumor growth was significantly inhibited at four weeks in animals vaccinated two weeks prior to tumor cell inoculation as compared to those given an empty vector control (1371 ± 420 mm(3) versus 405 ± 139 mm(3); p < 0.01). Mice vaccinated one week prior to tumor challenge also displayed significant reduction in tumor volume (1227 ± 406 mm(3) versus 309 ± 173 mm(3); p < 0.01). The vaccine had no effect when administered concurrently with tumor challenge, or after tumors were established. Flow cytometry showed reduced mesothelin expression in large tumors, as well as tumor-associated immunosuppression due to increased myeloid derived suppressor cells (MDSCs). These factors may have limited vaccine efficacy for advanced disease. Surgical cytoreduction of established tumors restored the antitumor potency of the therapeutic vaccine, with significantly reduced tumor burden at post-operative day 18 (397 ± 103 mm(3) versus 1047 ± 258 mm(3); p < 0.01). We found that surgery reduced MDSCs to levels comparable to those in tumor-naïve mice. This study demonstrates that cytoreduction surgery restores the efficacy of cancer vaccines for MPM by reducing tumor-related immunosuppression that impairs immunotherapy.


Neoplasia | 2012

Vascular Endothelial-Targeted Therapy Combined with Cytotoxic Chemotherapy Induces Inflammatory Intratumoral Infiltrates and Inhibits Tumor Relapses after Surgery

Brendan F. Judy; Louis A. Aliperti; Jarrod D. Predina; Daniel Levine; Veena Kapoor; Philip E. Thorpe; Steven M. Albelda; Sunil Singhal

Collaboration


Dive into the Brendan F. Judy's collaboration.

Top Co-Authors

Avatar

Sunil Singhal

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jarrod D. Predina

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Veena Kapoor

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Steven M. Albelda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Louis A. Aliperti

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jon G. Quatromoni

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zvi G. Fridlender

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Aaron Blouin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brian Madajewski

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge