Network


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

Hotspot


Dive into the research topics where Kevin F. Staveley-O’Carroll is active.

Publication


Featured researches published by Kevin F. Staveley-O’Carroll.


Journal of Immunology | 2003

In Vivo Ligation of CD40 Enhances Priming Against the Endogenous Tumor Antigen and Promotes CD8+ T Cell Effector Function in SV40 T Antigen Transgenic Mice

Kevin F. Staveley-O’Carroll; Todd D. Schell; Marcela Jimenez; Lawrence M. Mylin; M. Judith Tevethia; Stephen P. Schoenberger; Satvir S. Tevethia

The ability to initiate and sustain CD8+ T cell responses to tumors in vivo is hindered by the development of peripheral T cell tolerance against tumor-associated Ags. Approaches that counter the onset of T cell tolerance may preserve a pool of potentially tumor-reactive CD8+ T cells. Administration of agonist Ab to the CD40 molecule, expressed on APCs, can enhance immunization approaches targeting T lymphocytes in an otherwise tolerance-prone environment. In this report, the effects of anti-CD40 administration on priming of naive CD8+ T cells against an endogenous tumor Ag were investigated. Line 501 mice express the SV40 large T Ag oncoprotein as a transgene from the α-amylase promoter, resulting in the development of peripheral CD8+ T cell tolerance to the H-2-Db-restricted immunodominant epitope I of T Ag by 6 mo of age, before the appearance of osteosarcomas. We demonstrate that naive epitope I-specific TCR transgenic (TCR-I) T cells undergo peripheral tolerance following adoptive transfer into 6-mo-old 501 mice. In contrast, administration of agonistic anti-CD40 Ab led to increased expansion of TCR-I T cells in 501 mice, the acquisition of effector function by TCR-I T cells and the establishment of T cell memory. Importantly, this enhanced priming effect of anti-CD40 administration did not require immunization and was effective even if administered after naive TCR-I T cells had encountered the endogenous T Ag. Thus, anti-CD40 administration can block the onset of peripheral tolerance and enhance the recruitment of functionally competent effector T cells toward an endogenous tumor Ag.


Journal of Gastrointestinal Surgery | 2009

A Reduction in Delayed Gastric Emptying by Classic Pancreaticoduodenectomy with an Antecolic Gastrojejunal Anastomosis and a Retrogastric Omental Patch

Mehrdad Nikfarjam; Eric T. Kimchi; Niraj J. Gusani; Syed Shah; Mandeep Sehmbey; Serene Shereef; Kevin F. Staveley-O’Carroll

BackgroundDelayed gastric emptying (DGE) continues to be a major cause of morbidity following pancreaticoduodenectomy (PD). A change in the method of reconstruction following PD was instituted in an attempt to reduce the incidence DGE.MethodsPatients undergoing PD from January 2002 to December 2008 were reviewed and outcomes determined. Pylorus-preserving pancreaticoduodenectomy (PPPD) with a retrocolic duodenojejunal anastomosis (n = 79) or a classic PD with a retrocolic gastrojejunostomy (n = 36) was performed prior to January 2008. Thereafter, a classic PD with an antecolic gastrojejunal anastomosis and placement of a retrogastric vascular omental patch was undertaken (n = 36).ResultsA statistically significant decrease in DGE was noted in the antecolic group compared to the entire retrocolic group (14% vs 40%; p = 0.004) and compared to patients treated by classic PD with a retrocolic anastomosis alone (14% vs 39%; p = 0.016). On multivariate analysis, the only modifiable factor associated with reduced DGE was the antecolic technique with an omental patch, odds ratio (OR) 0.3 (confidence interval (CI) 0.1–0.8) p = 0.022. Male gender was associated with an increased risk of DGE with OR 2.3 (CI 1.1–4.8) p = 0.026.ConclusionA classic PD combined with an antecolic anastomosis and retrogastric vascular omental patch results in a significant reduction in DGE.


PLOS ONE | 2012

Circulating Tumor Cells in Melanoma Patients

Gary A. Clawson; Eric T. Kimchi; Susan D. Patrick; Ping Xin; Ramdane Harouaka; Siyang Zheng; Arthur Berg; Todd D. Schell; Kevin F. Staveley-O’Carroll; Rogerio I. Neves; Paul J. Mosca; Diane Thiboutot

Circulating tumor cells (CTCs) are of recognized importance for diagnosis and prognosis of cancer patients. With melanoma, most studies do not show any clear relationship between CTC levels and stage of disease. Here, CTCs were enriched (∼400X) from blood of melanoma patients using a simple centrifugation device (OncoQuick), and 4 melanocyte target RNAs (TYR, MLANA, MITF, and MIF) were quantified using QPCR. Approximately one-third of melanoma patients had elevated MIF and MLANA transcripts (p<0.0001 and p<0.001, respectively) compared with healthy controls. In contrast, healthy controls had uniformly higher levels of TYR and MITF than melanoma patients (p<0.0001). There was a marked shift of leukocytes into the CTC-enriched fractions (a 430% increase in RNA recovery, p<0.001), and no relationship between CTC levels and stage of disease was found. CTCs were captured on microfabricated filters and cultured. Captured melanoma CTCs were large cells, and consisted of 2 subpopulations, based on immunoreactivity. One subpopulation (∼50%) stained for both pan-cytokeratin (KRT) markers and the common leukocyte marker CD-45, whereas the second subpopulation stained for only KRT. Since similar cells are described in many cancers, we also examined blood from colorectal and pancreatic cancer patients. We observed analogous results, with most captured CTCs staining for both CD-45/KRT markers (and for the monocyte differentiation marker CD-14). Our results suggest that immature melanocyte-related cells (expressing TYR and MITF RNA) may circulate in healthy controls, although they are not readily detectable without considerable enrichment. Further, as early-stage melanomas develop, immature melanocyte migration into the blood is somehow curtailed, whereas a significant proportion of patients develop elevated CTC levels (based on MIF and MLANA RNAs). The nature of the captured CTCs is consistent with literature describing leukocyte/macrophage-tumor cell fusion hybrids, and their role in metastatic progression.


Journal of Surgical Research | 2004

Developing the young academic surgeon

Kevin F. Staveley-O’Carroll; Ming Pan; Andreas Meier; David Han; David W. McFadden; Wiley W. Souba

In the past, the process of developing the young academic surgeon was arguably less strategic, one that was often not deliberately managed and monitored, leading in some cases to academic drift and disillusionment. Once upon a time it was assumed that greatness was genetic and that the next triple threat would emerge when a pre-programmed set of genes was turned on. Today, as the complexities and vicissitudes of our work increase, it is practically impossible for even the most gifted young person to be successful without careful attention to career development. Faculty development must be deliberate and strategic--every junior faculty member is unique and will require a customized career development plan that is well thought out, linked to measurable goals, monitored routinely and buttressed by effective mentoring. This approach will require time and commitment--precious commodities that are in short supply as the demands on our time are only escalating. By recruiting the right people (those who fit with the organizations values and goals) and providing the right environment, we can optimize the growth and satisfaction of our young faculty and, in so doing, create departments that are leaders in carrying out our missions of research, education and patient care. We cannot afford to have our young people fail--it is simply too costly, both from a financial and a human perspective.


Bioorganic & Medicinal Chemistry | 2011

Synthesis and biological evaluation of a novel class of isatin analogs as dual inhibitors of tubulin polymerization and Akt pathway

Gowdahalli Krishnegowda; A. S. Prakasha Gowda; Hephzibah Rani S. Tagaram; Kevin F. Staveley-O’Carroll; Rosalyn B. Irby; Arun K. Sharma; Shantu Amin

A novel series of 5,7-dibromoisatin analogs were synthesized and evaluated for their cytotoxicities against four human cancer cell lines including colon HT29, breast MCF-7, lung A549 and melanoma UACC903. Analogs 6, 11 and 13 displayed good in vitro anticancer activity on the HT29 human colon cancer cell line in the 1 μM range. Analogs 5, 9 and 12, containing a selenocyanate group in the alkyl chain were the most promising compounds on the breast cancer MCF-7 cell line. Biological assays relating to apoptosis were performed to understand the mechanism of action of these analogs. Compounds 5 and 6 were found to inhibit tubulin polymerization to the same extent as the anticancer drug vinblastine sulfate, but compounds 11 and 13 inhibited significantly better than vinblastine. Further western blot analysis suggested that compound 6 at 2 μM reduced both levels and phosphorylation state of Akt. Compounds 11 and 13 at 1 μM caused reduced Akt protein levels and strongly suppressed the phosphorylation of Akt. Therefore, 11 and 13 were demonstrated as efficient dual inhibitors of both tubulin polymerization and the Akt pathway and good candidates for further study. More importantly, the strategy of microtubule and Akt dual inhibitors might be a promising direction for developing novel drugs for cancer.


Journal of General Internal Medicine | 2009

Cancer Survivorship: A New Challenge for Surgical and Medical Oncologists

Niraj J. Gusani; Jane R. Schubart; James Wise; Elana Farace; Michael J. Green; Yixing Jiang; Eric T. Kimchi; Kevin F. Staveley-O’Carroll

Increasingly, oncology practitioners are realizing that despite the “success” of cancer therapies, cancer survivors are facing previously unrecognized issues related to survivorship, including physical and psychosocial side-effects of the cancer and its treatment, second cancers, and practical and economic issues related to adjusting to life after their cancer diagnosis. In gastrointestinal cancers, traditionally, the medical and surgical oncologists charged with the care of the patient are not well-equipped to deal with these survivorship issues. At the Penn State Cancer Institute’s newly formed Gastrointestinal Cancer Survivorship Clinic, we provide a full array of services to optimize care through a multi-disciplinary approach. By utilizing a multi-disciplinary model with the primary medical and surgical oncologists spearheading the survivorship initiative and psychological and pastoral support as a priority, we hope to optimize cancer survivor care and patient quality of life.


Cancer Biology & Therapy | 2013

Circulating tumor cells are associated with diffuse spread in stage IV colorectal cancer patients.

Jussuf T. Kaifi; Miriam Kunkel; Junjia Zhu; David T. Dicker; Niraj J. Gusani; Zhaohai Yang; Nabeel Sarwani; Guangfu Li; Eric T. Kimchi; Kevin F. Staveley-O’Carroll; Wafik S. El-Deiry

Background Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the United States when combining both genders. Circulating tumor cells (CTCs) are a prognostic marker for stage IV CRC patients. We hypothesized that CTC quantity varies among stage IV CRC populations. Methods Blood (7.5 ml) was prospectively collected from 90 stage IV CRC patients. EpCAM+ CTCs were analyzed with the FDA-approved CellSearch® system. CRC tumors were immunohistochemically stained for EpCAM expression. Imaging and clinicopathological data were collected. Statistical analysis was performed using correlation analysis, Kruskal–Wallis, Fisher exact, and log-rank test. Results CTCs were detectable in 36/90 (40%) patients. Diffuse CRC metastases were associated with the highest CTC prevalence (24/40 [60%]), in contrast to limited lung (2/19 [11%]) or liver (10/31 [32%]) metastases (P = 0.027). The overall mean CTC number was 2.0 (range 0–56.3). The mean CTC number in patients with diffuse metastases was significantly higher (3.7 [SEM ± 1.7, range 0–56.3]) than with limited lung metastases (0.1 [± 0.1; range 0–1]) or liver metastases (0.9 [± 0.3, range 0–7.0]) (P = 0.001). CRC tumors were consistently expressing EpCAM. CTC numbers did not correlate with serum CEA levels or other routine clinical parameters (P = N.S.). Patients with diffuse metastases had the poorest overall survival (P = 0.0042). Conclusions CRC patients with diffuse metastases have the highest number of CTCs, in contrast to limited metastases to the liver or lungs. Future studies should correlate CTCs with recurrence patterns in patients with resected CRC lung or liver metastases to investigate whether CTCs represent micrometastatic disease causing early relapses.


Advances in Experimental Medicine and Biology | 2013

Impact of Genetic Targets on Cancer Therapy: Hepatocellular Cancer

Osama Hamed; Eric T. Kimchi; Mandeep Sehmbey; Niraj J. Gusani; Jussuf T. Kaifi; Kevin F. Staveley-O’Carroll

Understanding cancer at the genetic level had gained significant attention over the last decade since the human genome was first sequenced in 2001. For hepatocellular carcinoma (HCC) a number of genome-wide profiling studies have been published. These studies have provided us with gene sets, based on which we can now classify tumors and have an idea about the likely clinical outcomes. In addition to that, genomic profiling for HCC has provided us a better understanding of the carcinogenesis process and identifies key steps at multiple levels (i.e. Genetics, molecular pathways) that can be potential targets for treatment and prevention. Although still an incurable disease, unresectable HCC has one proven systemic therapy, sorafenib, and many under active investigation. With advancement in technology and understanding of hepatocarcinogenesis, scientists hope to provide true personalized treatment for this disease in the near future. In this review article we discuss advances in understanding genetics and pathogenesis of HCC and the currently available and ongoing trials for targeted therapies. These emerging therapies may guide the development of more effective treatments or possibly a cure for HCC.


Journal of Hepatology | 2017

Successful chemoimmunotherapy against hepatocellular cancer in a novel murine model.

Guangfu Li; Dai Liu; Timothy K. Cooper; Eric T. Kimchi; Xiaoqiang Qi; Diego M. Avella; Ningfei Li; Qing X. Yang; Mark Kester; C. Bart Rountree; Jussuf T. Kaifi; David J. Cole; Don C. Rockey; Todd D. Schell; Kevin F. Staveley-O’Carroll

BACKGROUND & AIMS We have established a clinically relevant animal model of hepatocellular cancer (HCC) in immune competent mice to elucidate the complex dialog between host immunity and tumors during HCC initiation and progression. Mechanistic findings have been leveraged to develop a clinically feasible anti-tumor chemoimmunotherapeutic strategy. METHODS Intraperitoneal injection of carbon tetrachloride and intrasplenic inoculation of oncogenic hepatocytes were combined to induce progressive HCCs in fibrotic livers of immunocompetent mice. Immunization and adoptive cell transfer (ACT) were used to dissect the tumor antigen-specific immune response. The ability of the tyrosine kinase inhibitor sunitinib to enhance immunotherapy in the setting of HCC was evaluated. RESULTS This new mouse model mimics human HCC and reflects its typical features. Tumor-antigen-specific CD8+ T cells maintained a naïve phenotype and remained responsive during early-stage tumor progression. Late tumor progression produced circulating tumor cells, tumor migration into draining lymph nodes, and profound exhaustion of tumor-antigen-specific CD8+ T cells associated with accumulation of programmed cell death protein 1 (PD-1)hi CD8+ T cells and regulatory T cells (Tregs). Sunitinib-mediated tumoricidal effect and Treg suppression synergized with antibody-mediated blockade of PD-1 to powerfully suppress tumor growth and activate anti-tumor immunity. CONCLUSION Treg accumulation and upregulation of PD-1 provide two independent mechanisms to induce profound immune tolerance in HCC. Chemoimmunotherapy using Food and Drug Administration-approved sunitinib with anti-PD-1 antibodies achieved significant tumor control, supporting translation of this approach for the treatment of HCC patients. LAY SUMMARY In the current study, we have established a clinically relevant mouse model which mimics human liver cancer. Using this unique model, we studied the response of the immune system to this aggressive cancer. Findings from this trial have led to the development of an innovative and clinically feasible chemoimmunotherapeutic strategy.


Science Advances | 2017

Antibody-mediated neutralization of soluble MIC significantly enhances CTLA4 blockade therapy

Jingyu Zhang; Dai Liu; Guangfu Li; Kevin F. Staveley-O’Carroll; Julie N. Graff; Zihai Li; Jennifer D. Wu

Cooperative therapeutic effect of anti-CTLA4 and anti-sMIC antibodies. Antibody therapy targeting cytotoxic T lymphocyte–associated antigen 4 (CTLA4) elicited survival benefits in cancer patients; however, the overall response rate is limited. In addition, anti-CTLA4 antibody therapy induces a high rate of immune-related adverse events. The underlying factors that may influence anti-CTLA4 antibody therapy are not well defined. We report the impact of a cancer-derived immune modulator, the human-soluble natural killer group 2D (NKG2D) ligand sMIC (soluble major histocompatibility complex I chain–related molecule), on the therapeutic outcome of anti-CTLA4 antibody using an MIC transgenic spontaneous TRAMP (transgenic adenocarcinoma of the mouse prostate)/MIC tumor model. Unexpectedly, animals with elevated serum sMIC (sMIChi) responded poorly to anti-CTLA4 antibody therapy, with significantly shortened survival due to increased lung metastasis. These sMIChi animals also developed colitis in response to anti-CTLA4 antibody therapy. Coadministration of an sMIC-neutralizing monoclonal antibody with the anti-CTLA4 antibody alleviated treatment-induced colitis in sMIChi animals and generated a cooperative antitumor therapeutic effect by synergistically augmenting innate and adoptive antitumor immune responses. Our findings imply that a new combination therapy could improve the clinical response to anti-CTLA4 antibody therapy. Our findings also suggest that prescreening cancer patients for serum sMIC may help in selecting candidates who will elicit a better response to anti-CTLA4 antibody therapy.

Collaboration


Dive into the Kevin F. Staveley-O’Carroll's collaboration.

Top Co-Authors

Avatar

Eric T. Kimchi

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jussuf T. Kaifi

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Niraj J. Gusani

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Guangfu Li

University of Missouri

View shared research outputs
Top Co-Authors

Avatar

Diego M. Avella

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Dai Liu

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Serene Shereef

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Todd D. Schell

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaoqiang Qi

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge