Network


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

Hotspot


Dive into the research topics where Sangeeta Goswami is active.

Publication


Featured researches published by Sangeeta Goswami.


Nature Medicine | 2007

Antielastin autoimmunity in tobacco smoking–induced emphysema

Seung Hyo Lee; Sangeeta Goswami; Ariel Grudo; Li Zhen Song; Venkata Bandi; Sheila Goodnight-White; Linda K. Green; Joan Hacken-Bitar; Joseph Huh; Faisal G. Bakaeen; Harvey O. Coxson; Sebastian Cogswell; Claudine Storness-Bliss; David B. Corry; Farrah Kheradmand

Chronic obstructive pulmonary disease and emphysema are common destructive inflammatory diseases that are leading causes of death worldwide. Here we show that emphysema is an autoimmune disease characterized by the presence of antielastin antibody and T-helper type 1 (TH1) responses, which correlate with emphysema severity. These findings link emphysema to adaptive immunity against a specific lung antigen and suggest the potential for autoimmune pathology of other elastin-rich tissues such as the arteries and skin of smokers.


Nature Communications | 2014

Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression

Limo Chen; Don L. Gibbons; Sangeeta Goswami; Maria Angelica Abdalla Cortez; Young Ho Ahn; Lauren Averett Byers; Xuejun Zhang; Xiaohui Yi; David Dwyer; Wei Lin; Lixia Diao; Jing Wang; Jonathon D. Roybal; Mayuri Patel; Christin Ungewiss; David H. Peng; Scott Antonia; Melanie Mediavilla-Varela; Gordon Robertson; Steve Jones; Milind Suraokar; James Welsh; Baruch Erez; Ignacio I. Wistuba; Lieping Chen; Di Peng; Shanshan Wang; Stephen E. Ullrich; John V. Heymach; Jonathan M. Kurie

Immunosuppression of tumor-infiltrating lymphocytes (TIL) is a common feature of advanced cancer, but its biological basis has remained obscure. We demonstrate here a molecular link between epithelial-to-mesenchymal transition (EMT) and CD8+ TIL immunosuppression, two key drivers of cancer progression. We show that microRNA-200 (miR-200), a cell-autonomous suppressor of EMT and metastasis, targets PD-L1. Moreover, ZEB1, an EMT activator and transcriptional repressor of miR-200, relieves miR-200 repression of PD-L1 on tumor cells, leading to CD8+ T cell immunosuppression and metastasis. These findings are supported by robust correlations between the EMT score, miR-200 levels and PD-L1 expression in multiple human lung cancer datasets. In addition to revealing a link between EMT and T cell dysfunction, these findings also show that ZEB1 promotes metastasis through a heretofore unappreciated cell non-autonomous mechanism, and suggest that subgroups of patients in whom malignant progression is driven by EMT activators may respond to treatment with PD-L1 antagonists.


Nature Immunology | 2009

Divergent functions for airway epithelial matrix metalloproteinase 7 and retinoic acid in experimental asthma

Sangeeta Goswami; Pornpimon Angkasekwinai; Ming Shan; Kendra J. Greenlee; Wade T. Barranco; Sumanth Polikepahad; Alexander Seryshev; Li Zhen Song; D. Redding; Bhupinder Singh; Sanjiv Sur; Prescott G. Woodruff; Chen Dong; David B. Corry; Farrah Kheradmand

The innate immune response of airway epithelial cells to airborne allergens initiates the development of T cell responses that are central to allergic inflammation. Although proteinase allergens induce the expression of interleukin 25, we show here that epithelial matrix metalloproteinase 7 (MMP7) was expressed during asthma and was required for the maximum activity of interleukin 25 in promoting the differentiation of T helper type 2 cells. Allergen-challenged Mmp7−/− mice had less airway hyper-reactivity and production of allergic inflammatory cytokines and higher expression of retinal dehydrogenase 1. Inhibition of retinal dehydrogenase 1 restored the asthma phenotype of Mmp7−/− mice and inhibited the responses of lung regulatory T cells, whereas exogenous administration of retinoic acid attenuated the asthma phenotype. Thus, MMP7 coordinates allergic lung inflammation by activating interleukin 25 while simultaneously inhibiting retinoid-dependent development of regulatory T cells.


Cancer Research | 2013

CXCR2 expression in tumor cells is a poor prognostic factor and promotes invasion and metastasis in lung adenocarcinoma

Pierre Saintigny; Erminia Massarelli; Steven H. Lin; Young Ho Ahn; Yulong Chen; Sangeeta Goswami; Baruch Erez; Michael S. O'Reilly; Diane Liu; J. Jack Lee; Li Zhang; Yuan Ping; Carmen Behrens; Luisa M.Solis Soto; John V. Heymach; Edward S. Kim; Roy S. Herbst; Scott M. Lippman; Ignacio I. Wistuba; Waun Ki Hong; Jonathan M. Kurie; Ja Seok Koo

CXCR2 in non-small cell lung cancer (NSCLC) has been studied mainly in stromal cells and is known to increase tumor inflammation and angiogenesis. Here, we examined the prognostic importance of CXCR2 in NSCLC and the role of CXCR2 and its ligands in lung cancer cells. The effect of CXCR2 expression on tumor cells was studied using stable knockdown clones derived from a murine KRAS/p53-mutant lung adenocarcinoma cell line with high metastatic potential and an orthotopic syngeneic mouse model and in vitro using a CXCR2 small-molecule antagonist (SB225002). CXCR2 protein expression was analyzed in tumor cells from 262 NSCLC. Gene expression profiles for CXCR2 and its ligands (CXCR2 axis) were analyzed in 52 human NSCLC cell lines and 442 human lung adenocarcinomas. Methylation of CXCR2 axis promoters was determined in 70 human NSCLC cell lines. Invasion and metastasis were decreased in CXCR2 knockdown clones in vitro and in vivo. SB225002 decreased invasion in vitro. In lung adenocarcinomas, CXCR2 expression in tumor cells was associated with smoking and poor prognosis. CXCR2 axis gene expression profiles in human NSCLC cell lines and lung adenocarcinomas defined a cluster driven by CXCL5 and associated with smoking, poor prognosis, and RAS pathway activation. Expression of CXCL5 was regulated by promoter methylation. The CXCR2 axis may be an important target in smoking-related lung adenocarcinoma.


Cancer Journal | 2016

Immune Checkpoint Therapies in Prostate Cancer.

Sangeeta Goswami; Ana Aparicio; Sumit K. Subudhi

Advanced prostate cancer is the second leading cause of death from cancer in the United States. In the era of cancer immunotherapy, it was the first malignancy to demonstrate improved survival with a cancer-specific vaccine, thus proving that prostate cancer is an immune-responsive disease. However, the success with immune checkpoint therapies in metastatic prostate cancer has been limited to date with only a subset of patients experiencing clinical benefit. The relative lack of response could be attributed to patient selection based on clinical attributes and the tumor microenvironment. Here, we review the current data on immune checkpoint therapies in prostate cancer and propose future directions.


OncoImmunology | 2016

Growth and metastasis of lung adenocarcinoma is potentiated by BMP4-mediated immunosuppression

Limo Chen; Xiaohui Yi; Sangeeta Goswami; Young Ho Ahn; Jonathon D. Roybal; Yongbin Yang; Lixia Diao; Di Peng; David H. Peng; Jared J. Fradette; Jing Wang; Lauren Averett Byers; Jonathan M. Kurie; Stephen E. Ullrich; F. Xiao Feng Qin; Don L. Gibbons

ABSTRACT Cancer cells modulate the recruitment and function of inflammatory cells to create an immunosuppressive microenvironment that favors tumor growth and metastasis. However, the tumor-derived regulatory programs that promote intratumoral immunosuppression remain poorly defined. Here, we show in a KrasLA1/+p53R172HΔg/+-based mouse model that bone morphogenetic protein-4 (BMP4) augments the expression of the T cell co-inhibitory receptor ligand PD-L1 in the mesenchymal subset of lung cancer cells, leading to profound CD8+ T cell-mediated immunosuppression, producing tumor growth and metastasis. We previously reported in this model that BMP4 functions as a pro-tumorigenic factor regulated by miR-200 via GATA4/6. Thus, BMP4‐mediated immunosuppression is part of a larger miR‐200‐directed gene expression program in tumors that promotes tumor progression, which could have important implications for cancer treatment.


Cancer Research | 2016

Abstract 4026: Epigenetic changes in T cells in response to immune checkpoint blockade

Sangeeta Goswami; Jianfeng Chen; Hao Zhao; Xuejun Zhang; Padmanee Sharma

Immune checkpoint blockade therapy has led to clinical success and have established immunotherapy as one of the primary modality of cancer treatment. It leads to a durable response in around 20% of the patients (responders). Success of immune checkpoint blockade therapy relies on the increase of effector T cells and decrease of regulatory T cells in the tumor microenvironment. However, the underlying mechanism that govern the ratio of effector and regulatory T cells between responder and non-responder to checkpoint blockade is not understood. Understanding the process could reveal novel mechanisms to boost efficacy of immune checkpoint blockade. Differentiation of naive T cells into effector and regulatory phenotype requires lineage-specifying transcription factors and epigenetic modifications that allow appropriate repression or activation of gene transcription. One key epigenetic modification for T cell differentiation is trimethylation of lysine 27 on histone H3 (H3K27me3). Loss of H3K27me3 results in increased Th-1 plasticity whereas presence of H3K27me3 on Foxp3 locus is required to maintain the function of T-regulatory cells. We hypothesize that immune checkpoint blockade changes the epigenetic landscape in tumor infiltrating T cells that results in an effector phenotype and epigenetic modification of H3K27me3 could promote an anti-tumor immune microenvironment in tumor bearing mice.We showed that in-vivo inhibition of H3K27me3 in combination with anti-CTLA4 results in reduction of tumor size and also reduction in regulatory T cells in a B16-F10 melanoma mice model compared to anti-CTLA4 treatment alone, suggesting H3K27me3 inhibition could increase the efficacy of checkpoint blockade therapy. Future studies will aim to elucidate epigenetic changes in H3K27me3 and define an epigenetic signature as a result of immune checkpoint therapy by ChIP-sequencing. Data generated in this project will greatly enhance our understanding of epigenetic regulation of T cell differentiation is response to immune checkpoint blockade and aid identifying key changes that could be further modified to overcome resistance to immune checkpoint blockade in non-responders. Citation Format: Sangeeta Goswami, Jianfeng Chen, Hao Zhao, Xuejun Zhang, Padmanee Sharma. Epigenetic changes in T cells in response to immune checkpoint blockade. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4026.


The Prostate | 2017

Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer; Report From the 2016 Coffey–Holden Prostate Cancer Academy Meeting

Andrea K. Miyahira; Sameek Roychowdhury; Sangeeta Goswami; Joseph E. Ippolito; Saul J. Priceman; Colin C. Pritchard; Karen S. Sfanos; Sumit Kumar Subudhi; Jonathan W. Simons; Kenneth J. Pienta; Howard R. Soule

The 2016 Coffey–Holden Prostate Cancer Academy (CHPCA) Meeting, “Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer,” was held from June 23 to June 26, 2016, in Coronado, California.


Journal of Clinical Investigation | 2018

Modulation of EZH2 expression in T cells improves efficacy of anti–CTLA-4 therapy

Sangeeta Goswami; Irina Apostolou; Jan Zhang; Jill Skepner; Swetha Anandhan; Xuejun Zhang; Liangwen Xiong; Patrick Trojer; Ana Aparicio; Sumit K. Subudhi; James P. Allison; Hao Zhao; Padmanee Sharma

Enhancer of zeste homolog 2–mediated (EZH2-mediated) epigenetic regulation of T cell differentiation and Treg function has been described previously; however, the role of EZH2 in T cell–mediated antitumor immunity, especially in the context of immune checkpoint therapy, is not understood. Here, we showed that genetic depletion of EZH2 in Tregs (FoxP3creEZH2fl/fl mice) leads to robust antitumor immunity. In addition, pharmacological inhibition of EZH2 in human T cells using CPI-1205 elicited phenotypic and functional alterations of the Tregs and enhanced cytotoxic activity of Teffs. We observed that ipilimumab (anti–CTLA-4) increased EZH2 expression in peripheral T cells from treated patients. We hypothesized that inhibition of EZH2 expression in T cells would increase the effectiveness of anti–CTLA-4 therapy, which we tested in murine models. Collectively, our data demonstrated that modulating EZH2 expression in T cells can improve antitumor responses elicited by anti–CTLA-4 therapy, which provides a strong rationale for a combination trial of CPI-1205 plus ipilimumab.


Science | 2017

Genetic biomarker for cancer immunotherapy

Sangeeta Goswami; Padmanee Sharma

A tumor-associated defect can identify patients to receive immune checkpoint therapy T cells eliminate pathogens by recognizing foreign proteins that are expressed on the cell surface (antigens). T cell activation in response to antigen occurs for a controlled period of time and is stopped by the expression of immune checkpoint proteins (1). Allison and colleagues proposed that antibody blockade of these proteins would enable prolonged T cell responses against cancer cells (2). The preclinical and clinical data that emerged using antibodies against two immune checkpoint proteins, cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death receptor-1 (PD-1), led to a paradigm shift in oncology as the treatment of some patients with these drugs led to tumor regression and durable survival for more than a decade (3). But as more patients with various types of cancer have been treated with immune checkpoint therapies, an enduring problem is to identify which patients are likely to respond. On page 409 of this issue, Le et al. (4) report the response to anti-PD-1 therapy (pembrolizumab) in 12 different tumor types, and show that patients who responded to the agent had defects in a DNA damage response pathway called mismatch repair.

Collaboration


Dive into the Sangeeta Goswami's collaboration.

Top Co-Authors

Avatar

Jonathan M. Kurie

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Don L. Gibbons

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ignacio I. Wistuba

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jing Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John V. Heymach

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lauren Averett Byers

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lixia Diao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jonathon D. Roybal

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Limo Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Padmanee Sharma

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge