9 results on '"Harika Gundlapalli"'
Search Results
2. Table S2 from Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression
- Author
-
Alana L. Welm, Elvelyn Fernandez, Amanda Jiang, H. Atakan Ekiz, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Abstract
Table S2 - Genes in immune cell clusters
- Published
- 2023
3. Data from Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression
- Author
-
Alana L. Welm, Elvelyn Fernandez, Amanda Jiang, H. Atakan Ekiz, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Abstract
Immunotherapy has potential to prevent and treat metastatic breast cancer, but strategies to enhance immune-mediated killing of metastatic tumors are urgently needed. We report that a ligand-independent isoform of Ron kinase (SF-Ron) is a key target to enhance immune infiltration and eradicate metastatic tumors. Host-specific deletion of SF-Ron caused recruitment of lymphocytes to micrometastases, augmented tumor-specific T-cell responses, and nearly eliminated breast cancer metastasis in mice. Lack of host SF-Ron caused stem-like TCF1+ CD4+ T cells with type I differentiation potential to accumulate in metastases and prevent metastatic outgrowth. There was a corresponding increase in tumor-specific CD8+ T cells, which were also required to eliminate lung metastases. Treatment of mice with a Ron kinase inhibitor increased tumor-specific CD8+ T cells and protected from metastatic outgrowth. These data provide a strong preclinical rationale to pursue small-molecule Ron kinase inhibitors for the prevention and treatment of metastatic breast cancer.Significance:The discovery that SF-Ron promotes antitumor immune responses has significant clinical implications. Therapeutic antibodies targeting full-length Ron may not be effective for immunotherapy; poor efficacy of such antibodies in trials may be due to their inability to block SF-Ron. Our data warrant trials with inhibitors targeting SF-Ron in combination with immunotherapy.This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2023
4. Table S1 from Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression
- Author
-
Alana L. Welm, Elvelyn Fernandez, Amanda Jiang, H. Atakan Ekiz, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Abstract
Table S1 - List of antibodies and reagents
- Published
- 2023
5. Abstract PS17-24: Inhibition of short-form ron eliminates breast cancer metastases through an immune-mediated mechanism
- Author
-
Huseyin Atakan Ekiz, Alana L. Welm, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Cancer ,Immunosuppression ,Immunotherapy ,medicine.disease ,Metastatic breast cancer ,Receptor tyrosine kinase ,Metastasis ,Breast cancer ,Immune system ,Oncology ,medicine ,Cancer research ,biology.protein ,business - Abstract
Metastatic breast cancer is the overwhelming cause of breast cancer mortality and is still incurable. The rapid development of immunotherapy is an exciting new area of research in metastatic breast cancer. However, the extreme immunosuppressive tumor environment poses a major challenge. A better understanding of how the immune system can be harnessed against metastatic cancer is required to improve patient outcomes. We previously showed that expression of the receptor tyrosine kinase Ron in the host, rather than Ron's tumor expression, contributed to tumor-associated immunosuppression and duo inhibition of Ron and CTLA-4 significantly reduced metastatic outgrowth. However, the actual mechanism remains unclear. The present study provides evidence that the N-terminal truncated isoform, short-form Ron (SF-Ron), is the major contributor in suppressing the anti-tumor immune responses and promoting metastatic outgrowth. Genetic deletion of host SF-Ron nearly eliminated breast cancer metastasis in mice, lead to systemic immune-activation, increased recruitment of lymphocytes to the site of metastasis, and augmented tumor-specific T-cell responses. Lack of SF-Ron also leads to the accumulation of CD4+ T-cells in the metastatic lungs and endowed with anti-tumor potential. Importantly, mice treated with small molecule Ron kinase inhibitor that targets both Ron and SF-Ron, produced significantly higher, active, tumor-specific CD8+ T-cells. Our study indicates that blocking Ron, especially the SF-Ron, remodels the metastatic lung microenvironment to enhance anti-tumor immunity. This study sheds light on the potential non-redundant roles of full-length and SF-Ron isoforms in mediating breast cancer metastasis and anti-tumor immune responses; and highlights the relevance of combining Ron inhibitors with immunotherapeutic agents to potentially improve treatment efficacy for metastatic breast cancer patients. Citation Format: Shu-Chin Alicia Lai, Harika Gundlapalli, Huseyin A Ekiz, Alana L Welm. Inhibition of short-form ron eliminates breast cancer metastases through an immune-mediated mechanism [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS17-24.
- Published
- 2021
6. Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression
- Author
-
H. Atakan Ekiz, Amanda Jiang, Alana L. Welm, Elvelyn Fernandez, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,medicine.medical_treatment ,Breast Neoplasms ,Article ,Mice ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Immunosuppression Therapy ,Lung ,business.industry ,Kinase ,Receptor Protein-Tyrosine Kinases ,Immunosuppression ,Immunotherapy ,medicine.disease ,Metastatic breast cancer ,medicine.anatomical_structure ,Oncology ,Cancer research ,Female ,business ,CD8 - Abstract
Immunotherapy has potential to prevent and treat metastatic breast cancer, but strategies to enhance immune-mediated killing of metastatic tumors are urgently needed. We report that a ligand-independent isoform of Ron kinase (SF-Ron) is a key target to enhance immune infiltration and eradicate metastatic tumors. Host-specific deletion of SF-Ron caused recruitment of lymphocytes to micrometastases, augmented tumor-specific T-cell responses, and nearly eliminated breast cancer metastasis in mice. Lack of host SF-Ron caused stem-like TCF1+ CD4+ T cells with type I differentiation potential to accumulate in metastases and prevent metastatic outgrowth. There was a corresponding increase in tumor-specific CD8+ T cells, which were also required to eliminate lung metastases. Treatment of mice with a Ron kinase inhibitor increased tumor-specific CD8+ T cells and protected from metastatic outgrowth. These data provide a strong preclinical rationale to pursue small-molecule Ron kinase inhibitors for the prevention and treatment of metastatic breast cancer. Significance: The discovery that SF-Ron promotes antitumor immune responses has significant clinical implications. Therapeutic antibodies targeting full-length Ron may not be effective for immunotherapy; poor efficacy of such antibodies in trials may be due to their inability to block SF-Ron. Our data warrant trials with inhibitors targeting SF-Ron in combination with immunotherapy. This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2020
7. A pipeline for identification and validation of tumor-specific antigens in a mouse model of metastatic breast cancer
- Author
-
William H. Hildebrand, Harika Gundlapalli, Wei R. Chen, Hem R. Gurung, Curtis McMurtrey, Alana L. Welm, Christa I. DeVette, Ashley R. Hoover, and Shu-Chin Alicia Lai
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Tumor specific ,Epitopes, T-Lymphocyte ,Breast Neoplasms ,Mice, Inbred Strains ,MHC Class I ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Breast cancer ,breast cancer ,Cancer immunotherapy ,Antigen ,Antigens, Neoplasm ,medicine ,Immunology and Allergy ,Animals ,Humans ,Neoplasm Metastasis ,MMTV-PyMT ,RC254-282 ,peptide prediction ,Original Research ,business.industry ,Advanced stage ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,medicine.disease ,Metastatic breast cancer ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Tumor immunology ,Identification (biology) ,Female ,Immunologic diseases. Allergy ,business ,NetH2pan - Abstract
Cancer immunotherapy continues to make headway as a treatment for advanced stage tumors, revealing an urgent need to understand the fundamentals of anti-tumor immune responses. Noteworthy is a scarcity of data pertaining to the breadth and specificity of tumor-specific T cell responses in metastatic breast cancer. Autochthonous transgenic models of breast cancer display spontaneous metastasis in the FVB/NJ mouse strain, yet a lack of knowledge regarding tumor-bound MHC/peptide immune epitopes in this mouse model limits the characterization of tumor-specific T cell responses, and the mechanisms that regulate T cell responses in the metastatic setting. We recently generated the NetH2pan prediction tool for murine class I MHC ligands by building an FVB/NJ H-2q ligand database and combining it with public information from six other murine MHC alleles. Here, we deployed NetH2pan in combination with an advanced proteomics workflow to identify immunogenic T cell epitopes in the MMTV-PyMT transgenic model for metastatic breast cancer. Five unique MHC I/PyMT epitopes were identified. These tumor-specific epitopes were confirmed to be presented by the class I MHC of primary MMTV-PyMT tumors and their T cell immunogenicity was validated. Vaccination using a DNA construct encoding a truncated PyMT protein generated CD8 + T cell responses to these MHC class I/peptide complexes and prevented tumor development. In sum, we have established an MHC-ligand discovery pipeline in FVB/NJ mice, identified and tracked H-2Dq/PyMT neoantigen-specific T cells, and developed a vaccine that prevents tumor development in this metastatic model of breast cancer.
- Published
- 2019
8. Abstract LB-008: KRAS engages AGO2 to enhance cellular transformation
- Author
-
Gideon Bollag, Harika Gundlapalli, Sunita Shankar, Chandan Kumar-Sinha, Kevin Shannon, Sethuramasundaram Pitchiaya, Rohit Malik, Ari J. Firestone, Nils G. Walter, Anastasia K. Yocum, Arul M. Chinnaiyan, Vishal Kothari, Yasmine N. White, Xuhong Cao, Jeanne A. Stuckey, Saravana M. Dhanasekaran, and Yasuyuki Hosono
- Subjects
Cancer Research ,Effector ,Mutant ,Argonaute ,Biology ,medicine.disease_cause ,Cell biology ,RNA silencing ,Oncology ,medicine ,Gene silencing ,Small GTPase ,KRAS ,PI3K/AKT/mTOR pathway - Abstract
Oncogenic mutations in RAS provide a compelling yet intractable therapeutic target. Approximately 30% of all cancers harbor activating mutations in the RAS family of small GTPase proteins, making it one of the most common oncogenic aberrations in humans. Normal RAS proteins (H, K or N-RAS) localize to the inner cell membrane and transduce extracellular growth signals by cycling between an “active” GTP-bound state and “inactive” GDP-bound state. Our understanding of RAS biology is primarily from RAS protein-effector interactions that activate a variety of effectors at the plasma membrane like RAF/PI3K/RalGDS. Yet, targeting mutant RAS proteins or its effectors / pathways remains a challenging endeavor for treating RAS driven cancers. For a comprehensive identification of RAS interactors, we recently performed co-immunoprecipitation (Co-IP) Mass Spectrometric analysis of RAS immunoprecipitates from multiple cancer cell lines with differing KRAS mutation status. In all the cell lines studies, we uncovered an interaction between RAS and the core component of the RNA silencing machinery, Argonaute 2 (AGO2). Endogenously expressed RAS and AGO2 co-sediment and co-localize in intracellular membrane bound endoplasmic reticulum. AGO2 binds the Switch II region in KRAS, irrespective of GDP/GTP bound to RAS. Both endogenous and overexpressed mutant forms of KRAS, attenuate AGO2 related gene silencing function. Using NIH3T3 AGO2-/- cells, we demonstrate that the RAS-AGO2 interaction is required for maximal mutant KRAS expression and cellular transformation. Overall, our studies suggest that through its interaction with AGO2, RAS function extends well beyond its canonical role in intracellular signaling. We will present detailed characterization of the RAS-AGO2 interaction and its functional aspects that we have discovered so far. Citation Format: Sunita Shankar, Sethuramasundaram Pitchiaya, Rohit Malik, Vishal Kothari, Yasuyuki Hosono, Anastasia K. Yocum, Harika Gundlapalli, Yasmine White, Ari Firestone, Xuhong Cao, Saravana M. Dhanasekaran, Jeanne Stuckey, Gideon Bollag, Kevin Shannon, Nils Walter, Chandan Kumar-Sinha, Arul M. Chinnaiyan. KRAS engages AGO2 to enhance cellular transformation. [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 LB-008.
- Published
- 2016
9. Abstract LB-058: Novel interactions of the RAS oncoprotein
- Author
-
Sunita Shankar, Saravana M. Dhanasekaran, Xiaoju Wang, Matthew Shuler, June Escara-Wilke, Yasuyuki Hosono, Ari J. Firestone, Vishalakshi Krishnan, Arul M. Chinnaiyan, Vishal Kothari, Xuhong Cao, Sethuramasundaram Pitchiaya, Jeanne A. Stuckey, Kevin Shannon, Nils G. Walter, Anastasia K. Yocum, Yasmine White, Gideon Bollag, Rohit Malik, Harika Gundlapalli, Anton Poliakov, Krishnapriya Chinnaswamy, Chandan Kumar-Sinha, and Shanker Kalyana-Sundaram
- Subjects
Genetics ,MAPK/ERK pathway ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Oncology ,GTPase-activating protein ,Kinase ,Anti-apoptotic Ras signalling cascade ,Small GTPase ,GTPase ,HRAS ,Biology ,Cell biology - Abstract
Approximately 30% of all cancers harbor activating mutations in the RAS family of small GTPase proteins, making it one of the most common oncogenic aberrations in humans. Normal RAS proteins (H, K or N-RAS) localize to the inner cell membrane and transduce extracellular growth signals by cycling between an “active” GTP-bound state and “inactive” GDP-bound state, through interactions with various “GTPase activating proteins” (GAPs) that promote RAS mediated GTP hydrolysis. Oncogenic mutants of RAS lose their catalytic activity or association with the GAP proteins, resulting in constitutively active GTP-bound state that signals through direct interactions with effector kinases like RAF and PI3K and activate the MEK/ERK and/or Akt, leading to activation of hallmark cancer pathways including growth factor independence, uncontrolled cell proliferation, evasion of apoptosis and immune responses, increased metabolism as well as metastases. Although RAS is the most frequently mutated gene driving multifarious pathways of oncogenesis, our knowledge of protein interactions involving RAS proteins have been largely limited to RAS binding domains in RAF/PI3K/RalGDS. Targeting mutant RAS proteins or its direct effectors, or pathways activated by RAS effectors remains a challenging endeavor for treating RAS driven cancers. Towards the goal of a thorough understanding of RAS biology through a comprehensive identification of its interactors, we performed IP-Mass Spectrometric analysis of pan-RAS immunoprecipitates from multiple cell lines. Interestingly in our experiments, apart from the well-known interactor RAF, we found evidence of several novel RAS interacting proteins, including many with DNA and RNA binding motifs. Our study validates these findings through cell-free protein interaction analyses and explores the possible biological effects of these novel RAS interactions in mutant KRAS driven cellular transformation. Note: This abstract was not presented at the meeting. Citation Format: Sunita Shankar, Rohit Malik, Vishal Kothari, Yasuyuki Hosono, Sethuramasundaram Pitchiaya, Shanker Kalyana-Sundaram, Anastasia Yocum, June Escara-Wilke, Harika Gundlapalli, Krishnapriya Chinnaswamy, Matthew Shuler, Anton Poliakov, Xiaoju Wang, Vishalakshi Krishnan, Yasmine White, Ari Firestone, Xuhong Cao, Saravana M. Dhanasekaran, Jeanne Stuckey, Gideon Bollag, Kevin Shannon, Nils G. Walter, Chandan Kumar-Sinha, Arul Chinnaiyan. Novel interactions of the RAS oncoprotein. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr LB-058. doi:10.1158/1538-7445.AM2015-LB-058
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.