Network


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

Hotspot


Dive into the research topics where Giorgos C. Karakousis is active.

Publication


Featured researches published by Giorgos C. Karakousis.


Annals of Surgical Oncology | 2004

Mitotic rate as a predictor of sentinel lymph nodepositivity in patients with thin melanomas

Susan B. Kesmodel; Giorgos C. Karakousis; Jeffrey Botbyl; Robert J. Canter; Robert T. Lewis; Peter M. Wahl; Kyla P. Terhune; Abass Alavi; David E. Elder; Michael E. Ming; DuPont Guerry; Phyllis A. Gimotty; Douglas L. Fraker; Brian J. Czerniecki; Francis R. Spitz

BackgroundLymphatic mapping and sentinel lymphadenectomy (LM/SL) provide important prognostic information for patients with early-stage melanoma. Although the use of this technique in patients with thin melanomas (≤1.00 mm) is not routine, risk factors that may predict sentinel lymph node (SLN) positivity in this patient population are under investigation. We sought to determine whether mitotic rate (MR) is associated with SLN positivity in thin-melanoma patients and, therefore, whether it may be used to risk-stratify and select patients for LM/SL.MethodsClinical and histopathologic variables were reviewed for 181 patients with thin melanomas who underwent LM/SL from January 1996 through January 2004. Univariate and multivariate logistic regression analyses were performed to identify factors associated with SLN positivity. Risk groups were defined on the basis of the development of a classification tree.ResultsThe overall SLN positivity rate was 5%. All patients with positive SLNs had an MR of >0. By univariate analysis, MR and thickness were significant predictors of SLN positivity. The association between MR and SLN positivity remained significant controlling for each of the other variables evaluated. On the basis of a classification tree, patients with an MR >0 and tumor thickness ≥.76 mm were identified as a higher-risk group, with an SLN positivity rate of 12.3%.ConclusionsIn patients with thin melanomas, MR >0 seems to be a significant predictor of SLN positivity that may be used to risk-stratify and select patients for LM/SL. To confirm these results, the predictive value of MR for SLN positivity needs to be validated in other populations of thin-melanoma patients.


Journal of Clinical Investigation | 2014

Targeting ER stress–induced autophagy overcomes BRAF inhibitor resistance in melanoma

Xiao Hong Ma; Sheng Fu Piao; Souvik Dey; Quentin McAfee; Giorgos C. Karakousis; Jessie Villanueva; Lori S. Hart; Samuel M. Levi; Janice Hu; Gao Zhang; Rossitza Lazova; Vincent Klump; John M. Pawelek; Xiaowei Xu; Wei Xu; Lynn M. Schuchter; Michael A. Davies; Meenhard Herlyn; Jeffrey D. Winkler; Constantinos Koumenis; Ravi K. Amaravadi

Melanomas that result from mutations in the gene encoding BRAF often become resistant to BRAF inhibition (BRAFi), with multiple mechanisms contributing to resistance. While therapy-induced autophagy promotes resistance to a number of therapies, especially those that target PI3K/mTOR signaling, its role as an adaptive resistance mechanism to BRAFi is not well characterized. Using tumor biopsies from BRAF(V600E) melanoma patients treated either with BRAFi or with combined BRAF and MEK inhibition, we found that BRAFi-resistant tumors had increased levels of autophagy compared with baseline. Patients with higher levels of therapy-induced autophagy had drastically lower response rates to BRAFi and a shorter duration of progression-free survival. In BRAF(V600E) melanoma cell lines, BRAFi or BRAF/MEK inhibition induced cytoprotective autophagy, and autophagy inhibition enhanced BRAFi-induced cell death. Shortly after BRAF inhibitor treatment in melanoma cell lines, mutant BRAF bound the ER stress gatekeeper GRP78, which rapidly expanded the ER. Disassociation of GRP78 from the PKR-like ER-kinase (PERK) promoted a PERK-dependent ER stress response that subsequently activated cytoprotective autophagy. Combined BRAF and autophagy inhibition promoted tumor regression in BRAFi-resistant xenografts. These data identify a molecular pathway for drug resistance connecting BRAFi, the ER stress response, and autophagy and provide a rationale for combination approaches targeting this resistance pathway.


Nature | 2017

T-cell invigoration to tumour burden ratio associated with anti-PD-1 response

Alexander C. Huang; Michael A. Postow; Robert J. Orlowski; Rosemarie Mick; Bertram Bengsch; Sasikanth Manne; Wei Xu; Shannon Harmon; Josephine R. Giles; Brandon Wenz; Matthew Adamow; Deborah Kuk; Katherine S. Panageas; Cristina Carrera; Phillip Wong; Felix Quagliarello; Bradley Wubbenhorst; Kurt D’Andrea; Kristen E. Pauken; Ramin S. Herati; Ryan P. Staupe; Jason M. Schenkel; Suzanne McGettigan; Shawn Kothari; Sangeeth M. George; Robert H. Vonderheide; Ravi K. Amaravadi; Giorgos C. Karakousis; Lynn M. Schuchter; Xiaowei Xu

Despite the success of monotherapies based on blockade of programmed cell death 1 (PD-1) in human melanoma, most patients do not experience durable clinical benefit. Pre-existing T-cell infiltration and/or the presence of PD-L1 in tumours may be used as indicators of clinical response; however, blood-based profiling to understand the mechanisms of PD-1 blockade has not been widely explored. Here we use immune profiling of peripheral blood from patients with stage IV melanoma before and after treatment with the PD-1-targeting antibody pembrolizumab and identify pharmacodynamic changes in circulating exhausted-phenotype CD8 T cells (Tex cells). Most of the patients demonstrated an immunological response to pembrolizumab. Clinical failure in many patients was not solely due to an inability to induce immune reinvigoration, but rather resulted from an imbalance between T-cell reinvigoration and tumour burden. The magnitude of reinvigoration of circulating Tex cells determined in relation to pretreatment tumour burden correlated with clinical response. By focused profiling of a mechanistically relevant circulating T-cell subpopulation calibrated to pretreatment disease burden, we identify a clinically accessible potential on-treatment predictor of response to PD-1 blockade.


Journal of Clinical Investigation | 2013

Melanoma adapts to RAF/MEK inhibitors through FOXD3-mediated upregulation of ERBB3

Ethan V. Abel; Kevin J. Basile; Curtis H. Kugel; Agnieszka K. Witkiewicz; Kaitlyn Le; Ravi K. Amaravadi; Giorgos C. Karakousis; Xiaowei Xu; Wei Xu; Lynn M. Schuchter; Jason B. Lee; Adam Ertel; Paolo Fortina; Andrew E. Aplin

The mechanisms underlying adaptive resistance of melanoma to targeted therapies remain unclear. By combining ChIP sequencing with microarray-based gene profiling, we determined that ERBB3 is upregulated by FOXD3, a transcription factor that promotes resistance to RAF inhibitors in melanoma. Enhanced ERBB3 signaling promoted resistance to RAF pathway inhibitors in cultured melanoma cell lines and in mouse xenograft models. ERBB3 signaling was dependent on ERBB2; targeting ERBB2 with lapatinib in combination with the RAF inhibitor PLX4720 reduced tumor burden and extended latency of tumor regrowth in vivo versus PLX4720 alone. These results suggest that enhanced ERBB3 signaling may serve as a mechanism of adaptive resistance to RAF and MEK inhibitors in melanoma and that cotargeting this pathway may enhance the clinical efficacy and extend the therapeutic duration of RAF inhibitors.


Cancer Discovery | 2013

Hypoxia Induces Phenotypic Plasticity and Therapy Resistance in Melanoma via the Tyrosine Kinase Receptors ROR1 and ROR2

Michael P. O'Connell; Katie Marchbank; Marie R. Webster; Alexander Valiga; Amanpreet Kaur; Adina Vultur; Ling Li; Meenhard Herlyn; Jessie Villanueva; Qin Liu; Xiangfan Yin; Sandy Widura; Nivia Ruiz; Tura C. Camilli; Fred E. Indig; Keith T. Flaherty; Jennifer A. Wargo; Dennie T. Frederick; Zachary A. Cooper; Suresh Nair; Ravi K. Amaravadi; Lynn M. Schuchter; Giorgos C. Karakousis; Wei Xu; Xaiowei Xu; Ashani T. Weeraratna

UNLABELLED An emerging concept in melanoma biology is that of dynamic, adaptive phenotype switching, where cells switch from a highly proliferative, poorly invasive phenotype to a highly invasive, less proliferative one. This switch may hold significant implications not just for metastasis, but also for therapy resistance. We demonstrate that phenotype switching and subsequent resistance can be guided by changes in expression of receptors involved in the noncanonical Wnt5A signaling pathway, ROR1 and ROR2. ROR1 and ROR2 are inversely expressed in melanomas and negatively regulate each other. Furthermore, hypoxia initiates a shift of ROR1-positive melanomas to a more invasive, ROR2-positive phenotype. Notably, this receptor switch induces a 10-fold decrease in sensitivity to BRAF inhibitors. In patients with melanoma treated with the BRAF inhibitor vemurafenib, Wnt5A expression correlates with clinical response and therapy resistance. These data highlight the fact that mechanisms that guide metastatic progression may be linked to those that mediate therapy resistance. SIGNIFICANCE These data show for the fi rst time that a single signaling pathway, the Wnt signaling pathway, can effectively guide the phenotypic plasticity of tumor cells, when primed to do so by a hypoxic microenvironment. Importantly, this increased Wnt5A signaling can give rise to a subpopulation of highly invasive cells that are intrinsically less sensitive to novel therapies for melanoma, and targeting the Wnt5A/ROR2 axis could improve the efficacy and duration of response for patients with melanoma on vemurafenib.


Cell Reports | 2013

Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma

Jessie Villanueva; Jeffrey R. Infante; Clemens Krepler; Patricia Reyes-Uribe; Minu Samanta; Hsin-Yi Chen; Bin Li; Rolf Swoboda; Melissa Wilson; Adina Vultur; Mizuho Fukunaba-Kalabis; Bradley Wubbenhorst; Thomas Y. Chen; Qin Liu; Katrin Sproesser; Douglas J. DeMarini; Tona M. Gilmer; Anne-Marie Martin; Ronen Marmorstein; David C. Schultz; David W. Speicher; Giorgos C. Karakousis; Wei Xu; Ravi K. Amaravadi; Xiaowei Xu; Lynn M. Schuchter; Meenhard Herlyn; Katherine L. Nathanson

Although BRAF and MEK inhibitors have proven clinical benefits in melanoma, most patients develop resistance. We report a de novo MEK2-Q60P mutation and BRAF gain in a melanoma from a patient who progressed on the MEK inhibitor trametinib and did not respond to the BRAF inhibitor dabrafenib. We also identified the same MEK2-Q60P mutation along with BRAF amplification in a xenograft tumor derived from a second melanoma patient resistant to the combination of dabrafenib and trametinib. Melanoma cells chronically exposed to trametinib acquired concurrent MEK2-Q60P mutation and BRAF-V600E amplification, which conferred resistance to MEK and BRAF inhibitors. The resistant cells had sustained MAPK activation and persistent phosphorylation of S6K. A triple combination of dabrafenib, trametinib, and the PI3K/mTOR inhibitor GSK2126458 led to sustained tumor growth inhibition. Hence, concurrent genetic events that sustain MAPK signaling can underlie resistance to both BRAF and MEK inhibitors, requiring novel therapeutic strategies to overcome it.


Annals of Surgical Oncology | 2006

Predictors of Regional Nodal Disease in Patients With Thin Melanomas

Giorgos C. Karakousis; Phyllis A. Gimotty; Jeffrey Botbyl; Susan B. Kesmodel; David E. Elder; Rosalie Elenitsas; Michael E. Ming; DuPont Guerry; Douglas L. Fraker; Brian J. Czerniecki; Francis R. Spitz

BackgroundMost melanoma patients present with thin (≤1.0 mm) lesions. Indications for sentinel lymph node (SLN) biopsy are not well defined for this group. Previously, we reported an association between mitotic rate (MR) and SLN positivity in these patients. The study was limited by a relatively small sample size and low statistical power. In this study, we evaluated a large population of patients with thin melanoma from the pre-SLN era to identify predictors of regional nodal disease (RND) that may serve as a surrogate for SLN positivity.MethodsEight hundred eighty-two patients evaluated between 1972 and 1991 were included in the study. Univariate and multivariate regression analyses were performed by using clinical and histological data to identify factors associated with RND. A multivariate logistic regression model was developed and applied to the previously reported group of patients with thin melanomas who underwent SLN biopsy between 1996 and 2004 for validation.ResultsThirty-eight patients (4.3%) had evidence of RND. In the multivariate analysis, a MR >0, vertical growth phase (VGP), male sex, and ulceration were statistically significant predictors of RND. Patients at the highest risk according to a classification tree analysis (VGP and MR >0) had an RND rate of 11.9%. The regression model developed predicted well the SLN status in the validation sample.ConclusionsInvestigation of a large pre-SLN population identified MR >0, ulceration, VGP, and male sex as independently predictive of RND in patients with thin melanomas. These factors may help to identify subgroups of these patients that have clinically significant risks of SLN positivity.


Nature | 2016

sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance

Amanpreet Kaur; Marie R. Webster; Katie Marchbank; Reeti Behera; Abibatou Ndoye; Curtis H. Kugel; Vanessa Dang; Jessica Appleton; Michael P. O'Connell; Phil F. Cheng; Alexander Valiga; Rachel Morissette; Nazli B. McDonnell; Luigi Ferrucci; Andrew V. Kossenkov; Katrina Meeth; Hsin Yao Tang; Xiangfan Yin; William H. Wood; Elin Lehrmann; Kevin G. Becker; Keith T. Flaherty; Dennie T. Frederick; Jennifer A. Wargo; Zachary A. Cooper; Michael T. Tetzlaff; Courtney W. Hudgens; Katherine M. Aird; Rugang Zhang; Xiaowei Xu

Cancer is a disease of ageing. Clinically, aged cancer patients tend to have a poorer prognosis than young. This may be due to accumulated cellular damage, decreases in adaptive immunity, and chronic inflammation. However, the effects of the aged microenvironment on tumour progression have been largely unexplored. Since dermal fibroblasts can have profound impacts on melanoma progression, we examined whether age-related changes in dermal fibroblasts could drive melanoma metastasis and response to targeted therapy. Here we find that aged fibroblasts secrete a Wnt antagonist, sFRP2, which activates a multi-step signalling cascade in melanoma cells that results in a decrease in β-catenin and microphthalmia-associated transcription factor (MITF), and ultimately the loss of a key redox effector, APE1. Loss of APE1 attenuates the response of melanoma cells to DNA damage induced by reactive oxygen species, rendering the cells more resistant to targeted therapy (vemurafenib). Age-related increases in sFRP2 also augment both angiogenesis and metastasis of melanoma cells. These data provide an integrated view of how fibroblasts in the aged microenvironment contribute to tumour progression, offering new possibilities for the design of therapy for the elderly.


Cancer | 2015

The rise in metastasectomy across cancer types over the past decade

Edmund K. Bartlett; Kristina D. Simmons; Heather Wachtel; Robert E. Roses; Douglas L. Fraker; Rachel R. Kelz; Giorgos C. Karakousis

Although studies of metastasectomy have been limited primarily to institutional experiences, reports of favorable long‐term outcomes have generated increasing interest. In the current study, the authors attempted to define the national practice patterns in metastasectomy for 4 common malignancies with varying responsiveness to systemic therapy.


Journal of Clinical Investigation | 2016

Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors

Gao Zhang; Dennie T. Frederick; Lawrence Wu; Zhi Wei; Clemens Krepler; Satish Srinivasan; Young Chan Chae; Xiaowei Xu; Harry Choi; Elaida Dimwamwa; Omotayo Ope; Batool Shannan; Devraj Basu; Dongmei Zhang; Manti Guha; Min Xiao; Sergio Randell; Katrin Sproesser; Wei Xu; Jephrey Y. Liu; Giorgos C. Karakousis; Lynn M. Schuchter; Tara C. Gangadhar; Ravi K. Amaravadi; Mengnan Gu; Caiyue Xu; Abheek Ghosh; Weiting Xu; Tian Tian; Jie Zhang

Targeting multiple components of the MAPK pathway can prolong the survival of patients with BRAFV600E melanoma. This approach is not curative, as some BRAF-mutated melanoma cells are intrinsically resistant to MAPK inhibitors (MAPKi). At the systemic level, our knowledge of how signaling pathways underlie drug resistance needs to be further expanded. Here, we have shown that intrinsically resistant BRAF-mutated melanoma cells with a low basal level of mitochondrial biogenesis depend on this process to survive MAPKi. Intrinsically resistant cells exploited an integrated stress response, exhibited an increase in mitochondrial DNA content, and required oxidative phosphorylation to meet their bioenergetic needs. We determined that intrinsically resistant cells rely on the genes encoding TFAM, which controls mitochondrial genome replication and transcription, and TRAP1, which regulates mitochondrial protein folding. Therefore, we targeted mitochondrial biogenesis with a mitochondrium-targeted, small-molecule HSP90 inhibitor (Gamitrinib), which eradicated intrinsically resistant cells and augmented the efficacy of MAPKi by inducing mitochondrial dysfunction and inhibiting tumor bioenergetics. A subset of tumor biopsies from patients with disease progression despite MAPKi treatment showed increased mitochondrial biogenesis and tumor bioenergetics. A subset of acquired drug-resistant melanoma cell lines was sensitive to Gamitrinib. Our study establishes mitochondrial biogenesis, coupled with aberrant tumor bioenergetics, as a potential therapy escape mechanism and paves the way for a rationale-based combinatorial strategy to improve the efficacy of MAPKi.

Collaboration


Dive into the Giorgos C. Karakousis's collaboration.

Top Co-Authors

Avatar

Douglas L. Fraker

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rachel R. Kelz

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Edmund K. Bartlett

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Robert E. Roses

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Xiaowei Xu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Lynn M. Schuchter

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wei Xu

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Paul J. Zhang

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Sinnamon

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David E. Elder

Hospital of the University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge