11 results on '"Barzin Y. Nabet"'
Search Results
2. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance
- Author
-
Irene Li and Barzin Y. Nabet
- Subjects
Exosomes ,Tumor microenvironment ,Therapy resistance ,Biomarkers ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Exosomes are small extracellular vesicles that contain genetic material, proteins, and lipids. They function as potent signaling molecules between cancer cells and the surrounding cells that comprise the tumor microenvironment (TME). Exosomes derived from both tumor and stromal cells have been implicated in all stages of cancer progression and play an important role in therapy resistance. Moreover, due to their nature as mediators of cell-cell communication, they are integral to TME-dependent therapy resistance. In this review, we discuss current exosome isolation and profiling techniques and their role in TME interactions and therapy resistance. We also explore emerging clinical applications of both exosomes as biomarkers, direct therapeutic targets, and engineered nanocarriers. In order to fully understand the TME, careful interrogation of exosomes and their cargo is critical. This understanding is a promising avenue for the development of effective clinical applications.
- Published
- 2019
- Full Text
- View/download PDF
3. 528 Activated T cells engage IL-1 and TNFa-driven tumor inflammation to support immune checkpoint inhibitor resistance in inflammatory solid malignancies
- Author
-
Eun Ji Kim, Barzin Y Nabet, Nam Woo Cho, Sophia M Guldberg, Kamir J Hiam-Galvez, Jacqueline L Yee, Rachel DeBarge, Katherine Wai, Lauren S Levine, Naa Asheley Ashitey, and Matthew H Spitzer
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
4. 606 IMpower110: Tertiary lymphoid structures (TLS) and clinical outcomes in advanced non-small cell lung cancer (NSCLC) treated with first-line atezolizumab or chemotherapy
- Author
-
Robert Johnston, Hartmut Koeppen, Roy S Herbst, Kurt Schalper, Filippo De Marinis, Giuseppe Giaccone, Jacek Jassem, Marcus Ballinger, Barani Kumar Rajendran, David R Spigel, Miguel López de Rodas, Minu K Srivastava, Jennifer M Giltnane, Barzin Y Nabet, David S Shames, Velimir Gayevskiy, Vy Ma, Ivette Estay, Tien Hoang, and Reena Amin
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. 579-B TIGIT and PD-L1 co-blockade promotes clonal expansion of non-exhausted anti-tumour CD8+ T cells by facilitating costimulation
- Author
-
Yan Qu, Ira Mellman, Ellen Duong, Thomas D Wu, Eugene Y Chiang, Katherine Nutsch, Karl L Banta, Charles W Tran, Stephanie Mittman, Barzin Y Nabet, and Namrata S Patil
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
6. Short Diagnosis-to-Treatment Interval Is Associated With Higher Circulating Tumor DNA Levels in Diffuse Large B-Cell Lymphoma
- Author
-
Joanne Soo, Florian Scherer, Andrea Garofalo, Brian Sworder, Mark Roschewski, Jason R. Westin, Davide Rossi, Olivier Casasnovas, Michael C. Jin, Wyndham H. Wilson, Stefan Alig, Charles Macaulay, Michel Meignan, Ash A. Alizadeh, Mohammad Shahrokh Esfahani, David M. Kurtz, Ulrich Dührsen, Gianluca Gaidano, Alexander F.M. Craig, Maximilian Diehn, Barzin Y. Nabet, Christine Schmitz, and Andreas Hüttmann
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Treatment interval ,Medizin ,Circulating Tumor DNA ,Young Adult ,Text mining ,Internal medicine ,medicine ,Humans ,Aged ,Aged, 80 and over ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,Prognosis ,medicine.disease ,Lymphoma ,Clinical trial ,Circulating tumor DNA ,Female ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma - Abstract
PURPOSE Patients with Diffuse Large B-cell Lymphoma (DLBCL) in need of immediate therapy are largely under-represented in clinical trials. The diagnosis-to-treatment interval (DTI) has recently been described as a metric to quantify such patient selection bias, with short DTI being associated with adverse risk factors and inferior outcomes. Here, we characterized the relationships between DTI, circulating tumor DNA (ctDNA), conventional risk factors, and clinical outcomes, with the goal of defining objective disease metrics contributing to selection bias. PATIENTS AND METHODS We evaluated pretreatment ctDNA levels in 267 patients with DLBCL treated across multiple centers in Europe and the United States using Cancer Personalized Profiling by Deep Sequencing. Pretreatment ctDNA levels were correlated with DTI, total metabolic tumor volumes (TMTVs), the International Prognostic Index (IPI), and outcome. RESULTS Short DTI was associated with advanced-stage disease ( P < .001) and higher IPI ( P < .001). We also found an inverse correlation between DTI and TMTV ( RS = −0.37; P < .001). Similarly, pretreatment ctDNA levels were significantly associated with stage, IPI, and TMTV (all P < .001), demonstrating that both DTI and ctDNA reflect disease burden. Notably, patients with shorter DTI had higher pretreatment ctDNA levels ( P < .001). Pretreatment ctDNA levels predicted short DTI independent of the IPI ( P < .001). Although each risk factor was significantly associated with event-free survival in univariable analysis, ctDNA level was prognostic of event-free survival independent of DTI and IPI in multivariable Cox regression (ctDNA: hazard ratio, 1.5; 95% CI [1.2 to 2.0]; IPI: 1.1 [0.9 to 1.3]; −DTI: 1.1 [1.0 to 1.2]). CONCLUSION Short DTI largely reflects baseline tumor burden, which can be objectively measured using pretreatment ctDNA levels. Pretreatment ctDNA levels therefore have utility for quantifying and guarding against selection biases in prospective DLBCL clinical trials.
- Published
- 2021
7. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities
- Author
-
Carl M. Gay, C. Allison Stewart, Elizabeth M. Park, Lixia Diao, Sarah M. Groves, Simon Heeke, Barzin Y. Nabet, Junya Fujimoto, Luisa M. Solis, Wei Lu, Yuanxin Xi, Robert J. Cardnell, Qi Wang, Giulia Fabbri, Kasey R. Cargill, Natalie I. Vokes, Kavya Ramkumar, Bingnan Zhang, Carminia M. Della Corte, Paul Robson, Stephen G. Swisher, Jack A. Roth, Bonnie S. Glisson, David S. Shames, Ignacio I. Wistuba, Jing Wang, Vito Quaranta, John Minna, John V. Heymach, Lauren Averett Byers, Gay, C. M., Stewart, C. A., Park, E. M., Diao, L., Groves, S. M., Heeke, S., Nabet, B. Y., Fujimoto, J., Solis, L. M., Lu, W., Xi, Y., Cardnell, R. J., Wang, Q., Fabbri, G., Cargill, K. R., Vokes, N. I., Ramkumar, K., Zhang, B., Della Corte, C. M., Robson, P., Swisher, S. G., Roth, J. A., Glisson, B. S., Shames, D. S., Wistuba, I. I., Wang, J., Quaranta, V., Minna, J., Heymach, J. V., and Byers, L. A.
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Prognosi ,medicine.medical_treatment ,Mice, Nude ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,neuroendocrine ,neoplasms ,Transcription factor ,Cisplatin ,Kinase ,Animal ,ASCL1 ,EMT ,Immunity ,SCLC ,POU2F3 ,Immunotherapy ,Gene signature ,Prognosis ,Small Cell Lung Carcinoma ,humanities ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,Lung Neoplasm ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,NEUROD1 ,intratumoral heterogeneity ,Cancer research ,Female ,medicine.drug ,Human ,Transcription Factors - Abstract
Despite molecular and clinical heterogeneity, small cell lung cancer (SCLC) is treated as a single entity with predictably poor results. Using tumor expression data and non-negative matrix factorization, we identify four SCLC subtypes defined largely by differential expression of transcription factors ASCL1, NEUROD1, and POU2F3 or low expression of all three transcription factor signatures accompanied by an Inflamed gene signature (SCLC-A, N, P, and I, respectively). SCLC-I experiences the greatest benefit from the addition of immunotherapy to chemotherapy, while the other subtypes each have distinct vulnerabilities, including to inhibitors of PARP, Aurora kinases, or BCL-2. Cisplatin treatment of SCLC-A patient-derived xenografts induces intratumoral shifts toward SCLC-I, supporting subtype switching as a mechanism of acquired platinum resistance. We propose that matching baseline tumor subtype to therapy, as well as manipulating subtype switching on therapy, may enhance depth and duration of response for SCLC patients. Gay et al. provide a classification for four subtypes of small cell lung cancer, each with unique molecular features and therapeutic vulnerabilities. An inflamed, mesenchymal subtype predicts benefit with the addition of immunotherapy to chemotherapy. Intratumoral switching between chemosensitive and chemoresistant subtypes accompanies therapeutic resistance.
- Published
- 2021
8. KEAP1/NFE2L2 mutations predict lung cancer radiation resistance that can be targeted by glutaminase inhibition
- Author
-
David M. Kurtz, Barzin Y. Nabet, Christian A. Kunder, Billy W. Loo, Joseph B. Shrager, Everett J. Moding, Natalie S. Lui, Sukhmani K. Padda, Jacob J. Chabon, Ryan B. Ko, Young-Jun Jeon, Michael Xiang, Kavitha Ramchandran, Emily G. Hamilton, Heather A. Wakelee, Peter G. Maxim, Mark F. Berry, Susie Grant Owen, Maximilian Diehn, Christopher H. Yoo, Joel W. Neal, Leah M. Backhus, Millie Das, Michael S. Binkley, June Ho Shin, Henning Stehr, Siyeon Rhee, Linda Gojenola, Diego Almanza, Ash A. Alizadeh, and Monica Nesselbush
- Subjects
0301 basic medicine ,business.industry ,Glutaminase ,DNA damage ,medicine.medical_treatment ,medicine.disease ,NFE2L2 ,Article ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Radioresistance ,Cancer research ,Medicine ,business ,Lung cancer ,Genotyping ,Gene - Abstract
Tumor genotyping is not routinely performed in localized non–small cell lung cancer (NSCLC) due to lack of associations of mutations with outcome. Here, we analyze 232 consecutive patients with localized NSCLC and demonstrate that KEAP1 and NFE2L2 mutations are predictive of high rates of local recurrence (LR) after radiotherapy but not surgery. Half of LRs occurred in tumors with KEAP1/NFE2L2 mutations, indicating that they are major molecular drivers of clinical radioresistance. Next, we functionally evaluate KEAP1/NFE2L2 mutations in our radiotherapy cohort and demonstrate that only pathogenic mutations are associated with radioresistance. Furthermore, expression of NFE2L2 target genes does not predict LR, underscoring the utility of tumor genotyping. Finally, we show that glutaminase inhibition preferentially radiosensitizes KEAP1-mutant cells via depletion of glutathione and increased radiation-induced DNA damage. Our findings suggest that genotyping for KEAP1/NFE2L2 mutations could facilitate treatment personalization and provide a potential strategy for overcoming radioresistance conferred by these mutations. Significance: This study shows that mutations in KEAP1 and NFE2L2 predict for LR after radiotherapy but not surgery in patients with NSCLC. Approximately half of all LRs are associated with these mutations and glutaminase inhibition may allow personalized radiosensitization of KEAP1/NFE2L2-mutant tumors. This article is highlighted in the In This Issue feature, p. 1775
- Published
- 2020
9. Circulating tumor DNA analysis to assess risk of progression after long-term response to PD-(L)1 blockade in NSCLC
- Author
-
Isabel Ruth Preeshagul, Mark G. Kris, Hyejin Choi, Henning Stehr, Andrew J. Plodkowski, Hira Rizvi, Christopher H. Yoo, Megan Tenet, Barzin Y. Nabet, Chih Long Liu, Charles M. Rudin, Joel W. Neal, Angela B. Hui, Sukhmani K. Padda, Jacob J. Chabon, Ash A. Alizadeh, Linda Gojenola, Jennifer L. Sauter, Diego Almanza, Mohsen Abu-Akeel, Jamie E. Chaft, Jia Luo, Mark Dunphy, Kathryn C. Arbour, Matthew D. Hellmann, Rene F. Bonilla, Maximilian Diehn, Everett J. Moding, Heather A. Wakelee, Bob T. Li, Rocio Perez Johnston, Daniel K. Wells, Aadel A. Chaudhuri, Taha Merghoub, and Ryan B. Ko
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Drug-Related Side Effects and Adverse Reactions ,Deep sequencing ,Article ,B7-H1 Antigen ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Biomarkers, Tumor ,Humans ,In patient ,business.industry ,Prognosis ,Minimal residual disease ,Immune checkpoint ,Blockade ,Long term response ,030104 developmental biology ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Disease Progression ,Non small cell ,business ,Follow-Up Studies - Abstract
Purpose: Treatment with PD-(L)1 blockade can produce remarkably durable responses in patients with non–small cell lung cancer (NSCLC). However, a significant fraction of long-term responders ultimately progress and predictors of late progression are unknown. We hypothesized that circulating tumor DNA (ctDNA) analysis of long-term responders to PD-(L)1 blockade may differentiate those who will achieve ongoing benefit from those at risk of eventual progression. Experimental Design: In patients with advanced NSCLC achieving long-term benefit from PD-(L)1 blockade (progression-free survival ≥ 12 months), plasma was collected at a surveillance timepoint late during/after treatment to interrogate ctDNA by Cancer Personalized Profiling by Deep Sequencing. Tumor tissue was available for 24 patients and was profiled by whole-exome sequencing (n = 18) or by targeted sequencing (n = 6). Results: Thirty-one patients with NSCLC with long-term benefit to PD-(L)1 blockade were identified, and ctDNA was analyzed in surveillance blood samples collected at a median of 26.7 months after initiation of therapy. Nine patients also had baseline plasma samples available, and all had detectable ctDNA prior to therapy initiation. At the surveillance timepoint, 27 patients had undetectable ctDNA and 25 (93%) have remained progression-free; in contrast, all 4 patients with detectable ctDNA eventually progressed [Fisher P < 0.0001; positive predictive value = 1, 95% confidence interval (CI), 0.51–1; negative predictive value = 0.93 (95% CI, 0.80–0.99)]. Conclusions: ctDNA analysis can noninvasively identify minimal residual disease in patients with long-term responses to PD-(L)1 blockade and predict the risk of eventual progression. If validated, ctDNA surveillance may facilitate personalization of the duration of immune checkpoint blockade and enable early intervention in patients at high risk for progression.
- Published
- 2020
10. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance
- Author
-
Barzin Y. Nabet and Irene Li
- Subjects
0301 basic medicine ,Cancer Research ,Cell signaling ,Stromal cell ,Review ,Cell Communication ,Biology ,Exosomes ,Exosome ,lcsh:RC254-282 ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Neoplasms ,Biomarkers, Tumor ,Humans ,CTLA-4 Antigen ,Treatment resistance ,Tumor microenvironment ,Drug Carriers ,Therapy resistance ,Endothelial Cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Antibodies, Neutralizing ,Microvesicles ,Killer Cells, Natural ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Disease Progression ,Molecular Medicine ,sense organs ,Immunotherapy ,Nanocarriers ,Stromal Cells ,Biomarkers - Abstract
Exosomes are small extracellular vesicles that contain genetic material, proteins, and lipids. They function as potent signaling molecules between cancer cells and the surrounding cells that comprise the tumor microenvironment (TME). Exosomes derived from both tumor and stromal cells have been implicated in all stages of cancer progression and play an important role in therapy resistance. Moreover, due to their nature as mediators of cell-cell communication, they are integral to TME-dependent therapy resistance. In this review, we discuss current exosome isolation and profiling techniques and their role in TME interactions and therapy resistance. We also explore emerging clinical applications of both exosomes as biomarkers, direct therapeutic targets, and engineered nanocarriers. In order to fully understand the TME, careful interrogation of exosomes and their cargo is critical. This understanding is a promising avenue for the development of effective clinical applications.
- Published
- 2019
11. Global analysis of shared T cell specificities in human non-small cell lung cancer enables HLA inference and antigen discovery
- Author
-
Sukhmani K. Padda, Alexandre Reuben, Mark M. Davis, Alana McSween, Xinbo Yang, Daisuke Nishimiya, Patrick Tran, P. Andrew Futreal, Julie Wilhelmy, Diane Tseng, Irene S. Molina, Jalen Benson, Natalie S. Lui, Heather A. Wakelee, Irving L. Weissman, John V. Heymach, Barzin Y. Nabet, Mark F. Berry, Joel W. Neal, K. Christopher Garcia, Rebecca Richards, Leah M. Backhus, Vamsee Mallajosyula, Poul H. Sorensen, Crystal L. Mackall, Elena Sotillo, Dorota Klysz, Ignacio I. Wistuba, Shin Heng Chiou, David M. Louis, Chunlin Wang, Alberto Delaidelli, Rahul Sinha, Joseph B. Shrager, Steven A. Feldman, Louai Labanieh, Stephanie D. Conley, Maximilian Diehn, Jianjun Zhang, and Gerald J. Berry
- Subjects
0301 basic medicine ,Resource ,EntS ,Lung Neoplasms ,cross-reactivity ,T cell ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Immunology ,Epitopes, T-Lymphocyte ,T-Cell Antigen Receptor Specificity ,Human leukocyte antigen ,Computational biology ,Biology ,Cross Reactions ,NSCLC ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,EBV ,LMP2A ,Antigens, Neoplasm ,tumor-infiltrating lymphocyte ,Carcinoma, Non-Small-Cell Lung ,HLA-A2 Antigen ,medicine ,Immunology and Allergy ,cancer ,Humans ,TMEM161A ,Cells, Cultured ,Antigen Presentation ,Tumor-infiltrating lymphocytes ,T-cell receptor ,Cancer ,medicine.disease ,T cell specificity ,GLIPH2 ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Paratope ,T cell receptor repertoire ,TCR ,Algorithms ,Epitope Mapping ,Protein Binding - Abstract
Summary To identify disease-relevant T cell receptors (TCRs) with shared antigen specificity, we analyzed 778,938 TCRβ chain sequences from 178 non-small cell lung cancer patients using the GLIPH2 (grouping of lymphocyte interactions with paratope hotspots 2) algorithm. We identified over 66,000 shared specificity groups, of which 435 were clonally expanded and enriched in tumors compared to adjacent lung. The antigenic epitopes of one such tumor-enriched specificity group were identified using a yeast peptide-HLA A∗02:01 display library. These included a peptide from the epithelial protein TMEM161A, which is overexpressed in tumors and cross-reactive epitopes from Epstein-Barr virus and E. coli. Our findings suggest that this cross-reactivity may underlie the presence of virus-specific T cells in tumor infiltrates and that pathogen cross-reactivity may be a feature of multiple cancers. The approach and analytical pipelines generated in this work, as well as the specificity groups defined here, present a resource for understanding the T cell response in cancer., Graphical abstract, Highlights • The algorithm GLIPH2 enables analysis of shared TCR specificity and HLA prediction • Tumor-infiltrating T cells cross-react to EBV antigens and shared tumor antigens • EBV-specific T cells expanded in patients responding to immune checkpoint blockade • Cross-reactive CD8 T cells express GZMK, Chiou, Tseng, et al. analyze TCRβ chain sequences from 178 non-small cell lung cancer patients and identify shared specificity groups, which in turn enable antigen identification. One such antigenic epitope—a peptide from an epithelial protein—is cross-reactive to epitopes from Epstein-Barr virus and E. coli, suggesting that cross-reactivity may underlie the presence of pathogen-specific T cells in tumor infiltrates.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.