11 results on '"Osman, Iman"'
Search Results
2. Melanoma expression of matrix metalloproteinase-23 is associated with blunted tumor immunity and poor responses to immunotherapy.
- Author
-
Moogk, Duane, da Silva, Ines Pires, Ma, Michelle W., Friedman, Erica B., de Miera, Eleazar Vega-Saenz, Darvishian, Farbod, Scanlon, Patrick, Perez-Garcia, Arianne, Pavlick, Anna C., Bhardwaj, Nina, Christos, Paul J., Osman, Iman, and Krogsgaar, Michelle
- Subjects
MELANOMA ,GENE expression ,MATRIX metalloproteinases ,TUMOR immunology ,IMMUNOTHERAPY ,CYTOKINE genetics ,T cell receptors ,LYMPHOCYTES ,GENETICS - Abstract
Background Matrix metalloproteinase-23 (MMP-23) can block the voltage-gated potassium channel Kv1.3, whose function is important for sustained Ca
2+ signaling during T cell activation. MMP-23 may also alter T cell activity and phenotype through cleavage of proteins affecting cytokine and chemokine signaling. We therefore tested the hypothesis that MMP-23 can negatively regulate the anti-tumor T cell response in human melanoma. Methods We characterized MMP-23 expression in primary melanoma patients who received adjuvant immunotherapy. We examined the association of MMP-23 with the anti-tumor immune response - as assessed by the prevalence of tumor-infiltrating lymphocytes and Foxp3+ regulatory T cells. Further, we examined the association between MMP-23 expression and response to immunotherapy. Considering also an in trans mechanism, we examined the association of melanoma MMP-23 and melanoma Kv1.3 expression. Results Our data revealed an inverse association between primary melanoma MMP-23 expression and the anti-tumor T cell response, as demonstrated by decreased tumor-infiltrating lymphocytes (TIL) (P = 0.05), in particular brisk TILs (P = 0.04), and a trend towards an increased proportion of immunosuppressive Foxp3+ regulatory T cells (P = 0.07). High melanoma MMP-23 expression is also associated with recurrence in patients treated with immune biologics (P = 0.037) but not in those treated with vaccines (P = 0.64). Further, high melanoma MMP-23 expression is associated with shorter periods of progression-free survival for patients receiving immune biologics (P = 0.025). On the other hand, there is no relationship between melanoma MMP-23 and melanoma Kv1.3 expression (P = 0.27). Conclusions Our data support a role for MMP-23 as a potential immunosuppressive target in melanoma, as well as a possible biomarker for informing melanoma immunotherapies. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
3. Hedgehog Pathway Blockade Inhibits Melanoma Cell Growth in Vitro and in Vivo.
- Author
-
O'Reilly, Kathryn E., de Miera, Eleazar Vega-Saenz, Segura, Miguel F., Friedman, Erica, Poliseno, Laura, Sung Won Han, Judy Zhong, Zavadil, Jiri, Pavlick, Anna, Hernando, Eva, and Osman, Iman
- Subjects
HERICIUM erinaceus ,MELANOMA ,CELL lines ,XENOGRAFTS ,HUMAN cloning ,GENE expression - Abstract
Previous reports have demonstrated a role for hedgehog signaling in melanoma progression, prompting us to explore the therapeutic benefit of targeting this pathway in melanoma. We profiled a panel of human melanoma cell lines and control melanocytes for altered expression of hedgehog pathway members and determined the consequences of both genetic and pharmacological inhibition of the hedgehog pathway activator Smoothened (SMO) in melanoma, both in vitro and in vivo. We also examined the relationship between altered expression of hedgehog pathway mediators and survival in a well-characterized cohort of metastatic melanoma patients with prospectively collected follow up information. Studies revealed that over 40% of the melanoma cell lines examined harbored significantly elevated levels of the hedgehog pathway mediators SMO, GLI2, and PTCH1 compared to melanocytes (p < 0.05). SMO inhibition using siRNA and the small molecule inhibitor, NVP-LDE-225, suppressed melanoma growth in vitro, particularly in those cell lines with moderate SMO and GLI2 expression. NVP-LDE-225 also induced apoptosis in vitro and inhibited melanoma growth in a xenograft model. Gene expression data also revealed evidence of compensatory up-regulation of two other developmental pathways, Notch and WNT, in response to hedgehog pathway inhibition. Pharmacological and genetic SMO inhibition also downregulated genes involved in human embryonic stem cell pluripotency. Finally, increased SMO expression and decreased expression of the hedgehog pathway repressor GLI3 correlated with shorter post recurrence survival in metastatic melanoma patients. Our data demonstrate that hedgehog pathway inhibition might be a promising targeted therapy in appropriately selected metastatic melanoma patients. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
4. The Novel Gamma Secretase Inhibitor RO4929097 Reduces the Tumor Initiating Potential of Melanoma.
- Author
-
Huynh, Chanh, Poliseno, Laura, Segura, Miguel F., Medicherla, Ratna, Haimovic, Adele, Menendez, Silvia, Shulian Shang, Pavlick, Anna, Yongzhao Shao, Darvishian, Farbod, Boylan, John F., Osman, Iman, and Hernando, Eva
- Subjects
GENE expression ,SECRETASE inhibitors ,CANCER ,LABORATORY mice ,NOTCH genes ,CANCER invasiveness ,CELL lines ,XENOGRAFTS - Abstract
Several reports have demonstrated a role for aberrant NOTCH signaling in melanoma genesis and progression, prompting us to explore if targeting this pathway is a valid therapeutic approach against melanoma. We targeted NOTCH signaling using RO4929097, a novel inhibitor of gamma secretase, which is a key component of the enzymatic complex that cleaves and activates NOTCH. The effects of RO4929097 on the oncogenic and stem cell properties of a panel of melanoma cell lines were tested both in vitro and in vivo, using xenograft models. In human primary melanoma cell lines, RO4929097 decreased the levels of NOTCH transcriptional target HES1. This was accompanied by reduced proliferation and impaired ability to form colonies in soft agar and to organize in tridimensional spheres. Moreover, RO4929097 affected the growth of human primary melanoma xenograft in NOD/SCID/IL2gammaR-/- mice and inhibited subsequent tumor formation in a serial xenotransplantation model, suggesting that inhibition of NOTCH signaling suppresses the tumor initiating potential of melanoma cells. In addition, RO4929097 decreased tumor volume and blocked the invasive growth pattern of metastatic melanoma cell lines in vivo. Finally, increased gene expression of NOTCH signaling components correlated with shorter post recurrence survival in metastatic melanoma cases. Our data support NOTCH inhibition as a promising therapeutic strategy against melanoma. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
5. microRNA-214 contributes to melanoma tumour progression through suppression of TFAP2C.
- Author
-
Penna, Elisa, Orso, Francesca, Cimino, Daniela, Tenaglia, Enrico, Lembo, Antonio, Quaglino, Elena, Poliseno, Laura, Haimovic, Adele, Osella-Abate, Simona, De Pittà, Cristiano, Pinatel, Eva, Stadler, Michael B, Provero, Paolo, Bernengo, Maria Grazia, Osman, Iman, and Taverna, Daniela
- Subjects
MELANOMA ,TUMOR prognosis ,METASTASIS ,CANCER cells ,GENE expression ,NON-coding RNA ,CELL adhesion - Abstract
Malignant melanoma is fatal in its metastatic stage. It is therefore essential to unravel the molecular mechanisms that govern disease progression to metastasis. MicroRNAs (miRs) are endogenous non-coding RNAs involved in tumourigenesis. Using a melanoma progression model, we identified a novel pathway controlled by miR-214 that coordinates metastatic capability. Pathway components include TFAP2C, homologue of a well-established melanoma tumour suppressor, the adhesion receptor ITGA3 and multiple surface molecules. Modulation of miR-214 influences in vitro tumour cell movement and survival to anoikis as well as extravasation from blood vessels and lung metastasis formation in vivo. Considering that miR-214 is known to be highly expressed in human melanomas, our data suggest a critical role for this miRNA in disease progression and the establishment of distant metastases. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
6. Clinical relevance of SKP2 alterations in metastatic melanoma.
- Author
-
Rose, Amy E., Wang, Guimin, Hanniford, Douglas, Monni, Stefano, Tu, Ting, Shapiro, Richard L., Berman, Russell S., Pavlick, Anna C., Pagano, Michele, Darvishian, Farbod, Mazumdar, Madhu, Hernando, Eva, and Osman, Iman
- Subjects
PROTO-oncogenes ,MELANOMA ,METASTASIS ,GENE expression ,CELL lines - Abstract
In this study, we investigated the mechanism(s) of altered expression of protooncogene SKP2 in metastatic melanoma and its clinical relevance in patients with metastatic melanoma. The genomic status of SKP2 was assessed in cell lines by sequencing, single nucleotide polymorphism array, and genomic PCR. Copy number status was then evaluated for concordance with SKP2 mRNA and protein expression. SKP2 protein was further evaluated by immunohistochemistry in 93 human metastatic tissues. No mutations were identified in SKP2. Increased copy number at the SKP2 locus was observed in 6/14 (43%) metastatic cell lines and in 9/22 (41%) human metastatic tissues which was associated with overexpression of SKP2 protein. Overexpression of SKP2 protein in human tissues was associated with worse survival in a multivariate model controlling for the site of metastasis. Copy number gain is a major contributing mechanism of SKP2 overexpression in metastatic melanoma. Results may have implications for the development of therapeutics that target SKP2. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
7. Copy number and gene expression differences between African American and Caucasian American prostate cancer.
- Author
-
Rose, Amy E., Satagopan, Jaya M., Oddoux, Carole, Qin Zhou, Ruliang Xu, Olshen, Adam B., Yu, Jessie Z., Dash, Atreya, Jean-Gilles, Jerome, Reuter, Victor, Gerald, William L., Peng Lee, and Osman, Iman
- Subjects
PROSTATE cancer ,HEALTH of African Americans ,CAUCASIAN race ,GENE expression ,MEDICAL care - Abstract
Background: The goal of our study was to investigate the molecular underpinnings associated with the relatively aggressive clinical behavior of prostate cancer (PCa) in African American (AA) compared to Caucasian American (CA) patients using a genome-wide approach. Methods: AA and CA patients treated with radical prostatectomy (RP) were frequency matched for age at RP, Gleason grade, and tumor stage. Array-CGH (BAC SpectralChip2600) was used to identify genomic regions with significantly different DNA copy number between the groups. Gene expression profiling of the same set of tumors was also evaluated using Affymetrix HG-U133 Plus 2.0 arrays. Concordance between copy number alteration and gene expression was examined. A second aCGH analysis was performed in a larger validation cohort using an oligo-based platform (Agilent 244K). Results: BAC-based array identified 27 chromosomal regions with significantly different copy number changes between the AA and CA tumors in the first cohort (Fisher's exact test, P < 0.05). Copy number alterations in these 27 regions were also significantly associated with gene expression changes. aCGH performed in a larger, independent cohort of AA and CA tumors validated 4 of the 27 (15%) most significantly altered regions from the initial analysis (3q26, 5p15-p14, 14q32, and 16p11). Functional annotation of overlapping genes within the 4 validated regions of AA/CA DNA copy number changes revealed significant enrichment of genes related to immune response. Conclusions: Our data reveal molecular alterations at the level of gene expression and DNA copy number that are specific to African American and Caucasian prostate cancer and may be related to underlying differences in immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
8. Stromal Anti-Apoptotic Androgen Receptor Target Gene c-FLIP in Prostate Cancer.
- Author
-
Ye, Huihui, Li, Yirong, Melamed, Jonathan, Pearce, Patrice, Wei, Jianjun, Chiriboga, Luis, Wang, Zhengxin, Osman, Iman, and Lee, Peng
- Subjects
PROSTATE cancer & genetics ,CANCER cell proliferation ,ANDROGENS ,HORMONE receptors ,GENETIC regulation ,APOPTOSIS ,GENE expression ,IMMUNOHISTOCHEMISTRY - Abstract
Purpose: The tumor microenvironment significantly influences prostate cancer progression. Androgen receptor exerts its effect through downstream target genes to regulate prostate cancer cell proliferation. The c-FLIP gene was recently shown to be an androgen receptor target gene. c-FLIP is an inactive homologue of caspase-8 and, thus, it inhibits the death receptor mediated apoptosis pathway. c-FLIP over expression was shown to accelerate the progression of prostate cancer cells to androgen independence. We evaluated the role of c-FLIP expression in stromal cells in prostate cancer development. Materials and Methods: We examined c-FLIP expression in 53 androgen dependent and 21 androgen independent prostate cancer stromal cells by immunohistochemical analysis. The effects of c-FLIP over expression in stromal cells on the growth and invasion of LNCaP and PC3 prostate cancer cells were determined in indirect coculture systems. Results: At the androgen dependent stage the stromal c-FLIP level was increased in prostate cancer tissue. The expression level of stromal c-FLIP was associated with tumor differentiation. However, stromal c-FLIP expression was not increased in androgen independent human prostate cancer. c-FLIP over expression in stromal cells stimulated the growth and invasion of prostate cancer, including LNCaP and PC3 cells in vitro. Conclusions: These results indicate the over expression of stromal c-FLIP and its function for promoting prostate cancer growth and invasion. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
9. Germline immunomodulatory expression quantitative trait loci (ieQTLs) associated with immune-related toxicity from checkpoint inhibition.
- Author
-
Ferguson, Robert, Chat, Vylyny, Morales, Leah, Simpson, Danny, Monson, Kelsey R., Cohen, Elisheva, Zusin, Sarah, Madonna, Gabriele, Capone, Mariaelena, Simeone, Ester, Pavlick, Anna, Luke, Jason J., Gajewski, Thomas F., Osman, Iman, Ascierto, Paolo, Weber, Jeffrey, and Kirchhoff, Tomas
- Subjects
- *
DRUG efficacy , *IMMUNE checkpoint inhibitors , *B cells , *CONFIDENCE intervals , *MELANOMA , *IPILIMUMAB , *IMMUNOMODULATORS , *GENETIC variation , *ALLELES , *GENE expression , *RISK assessment , *GENOTYPES , *DESCRIPTIVE statistics , *DRUG side effects , *TUMOR markers , *ODDS ratio , *LONGITUDINAL method , *DRUG toxicity - Abstract
Immune checkpoint inhibition (ICI) has improved clinical outcomes for metastatic melanoma patients; however, 65–80% of patients treated with ICI experience immune-related adverse events (irAEs). Given the plausible link of irAEs with underlying host immunity, we explored whether germline genetic variants controlling the expression of 42 immunomodulatory genes were associated with the risk of irAEs in melanoma patients treated with the single-agent anti-CTLA-4 antibody ipilimumab (IPI). We identified 42 immunomodulatory expression quantitative trait loci (ieQTLs) most significantly associated with the expression of 382 immune-related genes. These germline variants were genotyped in IPI-treated melanoma patients, collected as part of a multi-institutional collaboration. We tested the association of ieQTLs with irAEs in a discovery cohort of 95 patients, followed by validation in an additional 97 patients. We found that the alternate allele of rs7036417, a variant linked to increased expression of SYK , was strongly associated with an increased risk of grade 3–4 toxicity [odds ratio (OR) = 7.46; 95% confidence interval (CI) = 2.65–21.03; p = 1.43E-04]. This variant was not associated with response (OR = 0.90; 95% CI = 0.37–2.21; p = 0.82). We report that rs7036417 is associated with increased risk of severe irAEs, independent of IPI efficacy. SYK plays an important role in B-cell/T-cell expansion, and increased pSYK has been reported in patients with autoimmune disease. The association between rs7036417 and IPI irAEs in our data suggests a role of SYK overexpression in irAE development. These findings support the hypothesis that inherited variation in immune-related pathways modulates ICI toxicity and suggests SYK as a possible future target for therapies to reduce irAEs. • The current biomarkers of ICI toxicity in melanoma are insufficient. • Germline variants in immunomodulatory genes (ieQTLs) modulate ICI toxicity. • 35 ieQTLs were tested for the association with IPI toxicity. • ieQTL variant, linked to increased SYK expression, may predict severe IPI toxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Histology-Specific MicroRNA Alterations in Melanoma.
- Author
-
Poliseno, Laura, Haimovic, Adele, Segura, Miguel F, Hanniford, Douglas, Christos, Paul J, Darvishian, Farbod, Wang, Jinhua, Shapiro, Richard L, Pavlick, Anna C, Berman, Russell S, Hernando, Eva, Zavadil, Jiri, and Osman, Iman
- Subjects
- *
HISTOLOGY , *NON-coding RNA , *MELANOMA , *GENOMICS , *CELL lines , *HEALTH outcome assessment , *GENE expression - Abstract
We examined the microRNA signature that distinguishes the most common melanoma histological subtypes, superficial spreading melanoma (SSM) and nodular melanoma (NM). We also investigated the mechanisms underlying the differential expression of histology-specific microRNAs. MicroRNA array performed on a training cohort of 82 primary melanoma tumors (26 SSM, 56 NM), and nine congenital nevi (CN) revealed 134 microRNAs differentially expressed between SSM and NM (P<0.05). Out of 134 microRNAs, 126 remained significant after controlling for thickness and 31 were expressed at a lower level in SSM compared with both NM and CN. For seven microRNAs (let-7g, miR-15a, miR-16, miR-138, miR-181a, miR-191, and miR-933), the downregulation was associated with selective genomic loss in SSM cell lines and primary tumors, but not in NM cell lines and primary tumors. The lower expression level of six out of seven microRNAs in SSM compared with NM was confirmed by real-time PCR on a subset of cases in the training cohort and validated in an independent cohort of 97 melanoma cases (38 SSM, 59 NM). Our data support a molecular classification in which SSM and NM are two molecularly distinct phenotypes. Therapeutic strategies that take into account subtype-specific alterations might improve the outcome of melanoma patients. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
11. miR-30b/30d Regulation of GalNAc Transferases Enhances Invasion and Immunosuppression during Metastasis
- Author
-
Gaziel-Sovran, Avital, Segura, Miguel F., Di Micco, Raffaella, Collins, Mary K., Hanniford, Douglas, Vega-Saenz de Miera, Eleazar, Rakus, John F., Dankert, John F., Shang, Shulian, Kerbel, Robert S., Bhardwaj, Nina, Shao, Yongzhao, Darvishian, Farbod, Zavadil, Jiri, Erlebacher, Adrian, Mahal, Lara K., Osman, Iman, and Hernando, Eva
- Subjects
- *
CANCER cells , *TRANSFERASES , *IMMUNOSUPPRESSION , *METASTASIS , *GENE expression , *CYTOKINES , *GENETIC regulation , *CANCER invasiveness - Abstract
Summary: To metastasize, a tumor cell must acquire abilities such as the capacity to colonize new tissue and evade immune surveillance. Recent evidence suggests that microRNAs can promote the evolution of malignant behaviors by regulating multiple targets. We performed a microRNA analysis of human melanoma, a highly invasive cancer, and found that miR-30b/30d upregulation correlates with stage, metastatic potential, shorter time to recurrence, and reduced overall survival. Ectopic expression of miR-30b/30d promoted the metastatic behavior of melanoma cells by directly targeting the GalNAc transferase GALNT7, resulted in increased synthesis of the immunosuppressive cytokine IL-10, and reduced immune cell activation and recruitment. These data support a key role of miR-30b/30d and GalNAc transferases in metastasis, by simultaneously promoting cellular invasion and immunosuppression. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.