Network


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

Hotspot


Dive into the research topics where Paolo Cotzia is active.

Publication


Featured researches published by Paolo Cotzia.


Laryngoscope | 2017

Metformin effects on head and neck squamous carcinoma microenvironment: window of opportunity trial

Joseph Curry; Jennifer Maria Johnson; Patrick Tassone; Marina Domingo Vidal; Diana Whitaker Menezes; John Sprandio; Mehri Mollaee; Paolo Cotzia; Ruth Birbe; Zhao Lin; Kurren S. Gill; Elizabeth Duddy; Tingting Zhan; Benjamin E. Leiby; Michelle L. Reyzer; David Cognetti; Adam Luginbuhl; Madalina Tuluc; Ubaldo E. Martinez-Outschoorn

The tumor microenvironment frequently displays abnormal cellular metabolism, which contributes to aggressive behavior. Metformin inhibits mitochondrial oxidative phosphorylation, altering metabolism. Though the mechanism is unclear, epidemiologic studies show an association between metformin use and improved outcomes in head and neck squamous cell carcinoma (HNSCC). We sought to determine if metformin alters metabolism and apoptosis in HNSCC tumors.


Cancer Research | 2017

RB Loss Promotes Prostate Cancer Metastasis

Chellappagounder Thangavel; Ettickan Boopathi; Yi Liu; Alex Haber; Adam Ertel; Anshul Bhardwaj; Sankar Addya; Noelle L. Williams; Stephen J. Ciment; Paolo Cotzia; Jeffry L. Dean; Adam E. Snook; Chris McNair; Matthew Price; James R. Hernandez; Shuang G. Zhao; Ruth Birbe; James B. McCarthy; Eva A. Turley; Kenneth J. Pienta; Felix Y. Feng; Adam P. Dicker; Karen E. Knudsen; Robert B. Den

RB loss occurs commonly in neoplasia but its contributions to advanced cancer have not been assessed directly. Here we show that RB loss in multiple murine models of cancer produces a prometastatic phenotype. Gene expression analyses showed that regulation of the cell motility receptor RHAMM by the RB/E2F pathway was critical for epithelial-mesenchymal transition, motility, and invasion by cancer cells. Genetic modulation or pharmacologic inhibition of RHAMM activity was sufficient and necessary for metastatic phenotypes induced by RB loss in prostate cancer. Mechanistic studies in this setting established that RHAMM stabilized F-actin polymerization by controlling ROCK signaling. Collectively, our findings show how RB loss drives metastatic capacity and highlight RHAMM as a candidate therapeutic target for treating advanced prostate cancer. Cancer Res; 77(4); 982-95. ©2016 AACR.


Seminars in Oncology | 2015

Mitochondrial Metabolism as a Treatment Target in Anaplastic Thyroid Cancer

Jennifer Maria Johnson; Stephen Y. Lai; Paolo Cotzia; David Cognetti; Adam Luginbuhl; Edmund A. Pribitkin; Tingting Zhan; Mehri Mollaee; Marina Domingo-Vidal; Yunyun Chen; Barbara G. Campling; Voichita Bar-Ad; Ruth Birbe; Madalina Tuluc; Ubaldo Martinez Outschoorn; Joseph Curry

Anaplastic thyroid cancer (ATC) is one of the most aggressive human cancers. Key signal transduction pathways that regulate mitochondrial metabolism are frequently altered in ATC. Our goal was to determine the mitochondrial metabolic phenotype of ATC by studying markers of mitochondrial metabolism, specifically monocarboxylate transporter 1 (MCT1) and translocase of the outer mitochondrial membrane member 20 (TOMM20). Staining patterns of MCT1 and TOMM20 in 35 human thyroid samples (15 ATC, 12 papillary thyroid cancer [PTC], and eight non-cancerous thyroid) and nine ATC mouse orthotopic xenografts were assessed by visual and Aperio digital scoring. Staining patterns of areas involved with cancer versus areas with no evidence of cancer were evaluated independently where available. MCT1 is highly expressed in human anaplastic thyroid cancer when compared to both non-cancerous thyroid tissues and papillary thyroid cancers (P<.001 for both). TOMM20 is also highly expressed in both ATC and PTC compared to non-cancerous thyroid tissue (P<.01 for both). High MCT1 and TOMM20 expression is also found in ATC mouse xenograft tumors compared to non-cancerous thyroid tissue (P<.001). These xenograft tumors have high (13)C- pyruvate uptake. ATC has metabolic features that distinguish it from PTC and non-cancerous thyroid tissue, including high expression of MCT1 and TOMM20. PTC has low expression of MCT1 and non-cancerous thyroid tissue has low expression of both MCT1 and TOMM20. This work suggests that MCT1 blockade may specifically target ATC cells presenting an opportunity for a new drug target.


Frontiers in Cell and Developmental Biology | 2017

MCT1 in Invasive Ductal Carcinoma: Monocarboxylate Metabolism and Aggressive Breast Cancer

Jennifer Maria Johnson; Paolo Cotzia; Roberto Fratamico; Lekha Mikkilineni; Jason Chen; Daniele Colombo; Mehri Mollaee; Diana Whitaker-Menezes; Marina Domingo-Vidal; Zhao Lin; Tingting Zhan; Madalina Tuluc; Juan P. Palazzo; Ruth Birbe; Ubaldo E. Martinez-Outschoorn

Introduction: Monocarboxylate transporter 1 (MCT1) is an importer of monocarboxylates such as lactate and pyruvate and a marker of mitochondrial metabolism. MCT1 is highly expressed in a subgroup of cancer cells to allow for catabolite uptake from the tumor microenvironment to support mitochondrial metabolism. We studied the protein expression of MCT1 in a broad group of breast invasive ductal carcinoma specimens to determine its association with breast cancer subtypes and outcomes. Methods: MCT1 expression was evaluated by immunohistochemistry on tissue micro-arrays (TMA) obtained through our tumor bank. Two hundred and fifty-seven cases were analyzed: 180 cases were estrogen receptor and/or progesterone receptor positive (ER+ and/or PR+), 62 cases were human epidermal growth factor receptor 2 positive (HER2+), and 56 cases were triple negative breast cancers (TNBC). MCT1 expression was quantified by digital pathology with Aperio software. The intensity of the staining was measured on a continuous scale (0-black to 255-bright white) using a co-localization algorithm. Statistical analysis was performed using a linear mixed model. Results: High MCT1 expression was more commonly found in TNBC compared to ER+ and/or PR+ and compared to HER-2+ (p < 0.001). Tumors with an in-situ component were less likely to stain strongly for MCT1 (p < 0.05). High nuclear grade was associated with higher MCT1 staining (p < 0.01). Higher T stage tumors were noted to have a higher expression of MCT1 (p < 0.05). High MCT1 staining in cancer cells was associated with shorter progression free survival, increased risk of recurrence, and larger size independent of TNBC status (p < 0.05). Conclusion: MCT1 expression, which is a marker of high catabolite uptake and mitochondrial metabolism, is associated with recurrence in breast invasive ductal carcinoma. MCT1 expression as quantified with digital image analysis may be useful as a prognostic biomarker and to design clinical trials using MCT1 inhibitors.


Seminars in Oncology | 2017

Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-Acetylcysteine in Breast Cancer

Daniela Monti; Federica Sotgia; Diana Whitaker-Menezes; Madalina Tuluc; Ruth Birbe; Adam C. Berger; Melissa Lazar; Paolo Cotzia; Rossitza Draganova-Tacheva; Zhao Lin; Marina Domingo-Vidal; Andrew B. Newberg; Michael P. Lisanti; Ubaldo E. Martinez-Outschoorn

BACKGROUND High oxidative stress as defined by hydroxyl and peroxyl activity is often found in the stroma of human breast cancers. Oxidative stress induces stromal catabolism, which promotes cancer aggressiveness. Stromal cells exposed to oxidative stress release catabolites such as lactate, which are up-taken by cancer cells to support mitochondrial oxidative phosphorylation. The transfer of catabolites between stromal and cancer cells leads to metabolic heterogeneity between these cells and increased cancer cell proliferation and reduced apoptosis in preclinical models. N-Acetylcysteine (NAC) is an antioxidant that reduces oxidative stress and reverses stromal catabolism and stromal-carcinoma cell metabolic heterogeneity, resulting in reduced proliferation and increased apoptosis of cancer cells in experimental models of breast cancer. The purpose of this clinical trial was to determine if NAC could reduce markers of stromal-cancer metabolic heterogeneity and markers of cancer cell aggressiveness in human breast cancer. METHODS Subjects with newly diagnosed stage 0 and I breast cancer who were not going to receive neoadjuvant therapy prior to surgical resection were treated with NAC before definitive surgery to assess intra-tumoral metabolic markers. NAC was administered once a week intravenously at a dose of 150 mg/kg and 600 mg twice daily orally on the days not receiving intravenous NAC. Histochemistry for the stromal metabolic markers monocarboxylate transporter 4 (MCT4) and caveolin-1 (CAV1) and the Ki67 proliferation assay and TUNEL apoptosis assay in carcinoma cells were performed in pre- and post-NAC specimens. RESULTS The range of days on NAC was 14-27 and the mean was 19 days. Post-treatment biopsies showed significant decrease in stromal MCT4 and reduced Ki67 in carcinoma cells. NAC did not significantly change stromal CAV1 and carcinoma TUNEL staining. NAC was well tolerated. CONCLUSIONS NAC as a single agent reduces MCT4 stromal expression, which is a marker of glycolysis in breast cancer with reduced carcinoma cell proliferation. This study suggests that modulating metabolism in the tumor microenvironment has the potential to impact breast cancer proliferation.


Laryngoscope | 2016

Multicompartment metabolism in papillary thyroid cancer

Joseph Curry; Patrick Tassone; Paolo Cotzia; John Sprandio; Adam Luginbuhl; David Cognetti; Mehri Mollaee; Marina Domingo-Vidal; Edmund A. Pribitkin; William M. Keane; Tingting Zhan; Ruth Birbe; Madalina Tuluc; Ubaldo E. Martinez-Outschoorn

In many cancers, varying regions within the tumor are often phenotypically heterogeneous, including their metabolic phenotype. Further, tumor regions can be metabolically compartmentalized, with metabolites transferred between compartments. When present, this metabolic coupling can promote aggressive behavior. Tumor metabolism in papillary thyroid cancer (PTC) is poorly characterized.


Seminars in Oncology | 2015

Parathyroid Hormone-Related Peptide–Linked Hypercalcemia in a Melanoma Patient Treated With Ipilimumab: Hormone Source and Clinical and Metabolic Correlates

Teresa Anne Mills; Marlana M. Orloff; Marina Domingo-Vidal; Paolo Cotzia; Ruth Birbe; Rossitza Draganova-Tacheva; Maria P. Martinez Cantarin; Madalina Tuluc; Ubaldo E. Martinez-Outschoorn

A patient diagnosed with metastatic melanoma developed the paraneoplastic syndrome of humoral hypercalcemia of malignancy and cachexia after receiving ipilumumab. The cause of the hypercalcemia was thought to be secondary to parathyroid hormone-related peptide (PTHrP) as plasma levels were found to be elevated. The patient underwent two tumor biopsies: at diagnosis (when calcium levels were normal) and upon development of hypercalcemia and cachexia. PTHrP expression was higher in melanoma cells when hypercalcemia had occurred than prior to its onset. Metabolic characterization of melanoma cells revealed that, with development of hypercalcemia, there was high expression of monocarboxylate transporter 1 (MCT1), which is the main importer of lactate and ketone bodies into cells. MCT1 is associated with high mitochondrial metabolism. Beta-galactosidase (β-GAL), a marker of senescence, had reduced expression in melanoma cells upon development of hypercalcemia compared to pre-hypercalcemia. In conclusion, PTHrP expression in melanoma is associated with cachexia, increased cancer cell lactate and ketone body import, high mitochondrial metabolism, and reduced senescence. Further studies are required to determine if PTHrP regulates cachexia, lactate and ketone body import, mitochondrial metabolism, and senescence in cancer cells.


Human Pathology | 2018

JAK2, PD-L1, and PD-L2 (9p24.1) Amplification in Metastatic Mucosal and Cutaneous Melanomas with Durable Response to Immunotherapy

Sounak Gupta; Chad M. Vanderbilt; Paolo Cotzia; Javier Arias Stella; Jason C. Chang; Ying-Bei Chen; Laura H. Tang; Deborah DeLair; Jinjuan Yao; Marc Ladanyi; Dara S. Ross

As immune checkpoint inhibitors are rapidly developing into the standard of care for patients with advanced melanoma, the value of diagnostic metrics to predict response to immunotherapy is steadily increasing. Next-generation sequencing-based parameters include tumor mutation burden (TMB) and genomic amplification of PD-L1/PD-L2/JAK2 at 9p24.1. At present, there are limited studies documenting response to checkpoint blockade in 9p24.1-amplified solid tumors. Herein, we have compared a cutaneous melanoma with a mucosal melanoma, both with 9p24.1 amplifications and durable response to immunotherapy. Although the cutaneous melanoma had a high TMB, the mucosal melanoma had a lower TMB compared with the mean TMB for all melanomas within the institutional clinical sequencing cohort. In summary, PD-L1/PD-L2/JAK2 amplification was associated with durable response to therapy for both cases, and this genomic signature is a potential valuable metric in predicting response to immunotherapy.


Genes, Chromosomes and Cancer | 2018

A novel group of spindle cell tumors defined by S100 and CD34 co-expression shows recurrent fusions involving RAF1, BRAF, and NTRK1/2 genes

Albert J. H. Suurmeijer; Brendan C. Dickson; David Swanson; Lei Zhang; Yun-Shao Sung; Paolo Cotzia; Christopher D. M. Fletcher; Cristina R. Antonescu

Tumors characterized by co‐expression of S100 and CD34, in the absence of SOX10, remain difficult to classify. Triggered by a few index cases with monomorphic cytomorphology and distinctive stromal and perivascular hyalinization, immunopositivity for S100 and CD34, and RAF1 and NTRK1 fusions, the authors undertook a systematic review of tumors with similar features. Most of the cases selected were previously diagnosed as low‐grade malignant peripheral nerve sheath tumors, while others were deemed unclassified. The tumors were studied with targeted RNA sequencing and/or FISH. A total of 25 cases (15 adults and 10 children) with kinase fusions were identified, including 8 cases involving RAF1, 2 BRAF, 14 NTRK1, and 1 NTRK2 gene rearrangements. Most tumors showed a monomorphic spindle cell proliferation with stromal and perivascular keloidal collagen, in a patternless architecture, with only occasional scattered pleomorphic or multinucleated cells. Most cases showed low cellularity, a low mitotic count, and absence of necrosis. Although a subset showed overlap with lipofibromatosis‐like neural tumors, the study group showed distinctive hyalinization and overt malignant features, such as highly cellular fascicular growth and primitive appearance. All tumors showed co‐expression of S100 and CD34, ranging from focal to diffuse. SOX10 was negative in all cases. NTRK1 immunohistochemistry showed high levels of expression in all tumors with NTRK1 gene rearrangements. H3K27me3 expression performed in a subset of cases was retained. These findings together with the recurrent gene fusions in RAF1, BRAF, and NTRK1/2 kinases suggest a distinct molecular tumor subtype with consistent S100 and CD34 immunoreactivity.


The American Journal of Surgical Pathology | 2018

NTRK Fusions Define a Novel Uterine Sarcoma Subtype With Features of Fibrosarcoma

Sarah Chiang; Paolo Cotzia; David M. Hyman; Alexander Drilon; William D. Tap; Lei Zhang; Jaclyn F. Hechtman; Denise Frosina; Achim A. Jungbluth; Rajmohan Murali; Kay J. Park; Robert A. Soslow; Esther Oliva; A. John Iafrate; Ryma Benayed; Marc Ladanyi; Cristina R. Antonescu

Collaboration


Dive into the Paolo Cotzia's collaboration.

Top Co-Authors

Avatar

Ruth Birbe

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Madalina Tuluc

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mehri Mollaee

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Tingting Zhan

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam Luginbuhl

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

David Cognetti

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph Curry

Thomas Jefferson University

View shared research outputs
Researchain Logo
Decentralizing Knowledge