11 results on '"Asha Dhanarajan"'
Search Results
2. Integrin subunit beta 8 contributes to lenvatinib resistance in HCC
- Author
-
Wei Hou, Bryan Bridgeman, Greg Malnassy, Xianzhong Ding, Scott J. Cotler, Asha Dhanarajan, and Wei Qiu
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Abstract Lenvatinib is a multikinase inhibitor approved as a first‐line therapy for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance is common, and the underlying mechanisms governing this resistance are largely unknown. In this study, we established two lenvatinib‐resistant (LR) HCC cell lines and identified integrin subunit beta 8 (ITGB8) as a critical contributor to lenvatinib resistance in HCC. The elevated expression of ITGB8 was observed in LR HCC cells. Furthermore, silencing of ITGB8 reversed lenvatinib resistance in vitro and in vivo, whereas ectopic expression of ITGB8 in lenvatinib‐sensitive parental HCC cells exhibited increased resistance to lenvatinib. Mechanistically, ITGB8 regulated lenvatinib resistance through an HSP90‐mediated stabilization of AKT and enhanced AKT signaling. In support of this model, either an AKT inhibitor MK‐2206 or an HSP90 inhibitor 17‐AAG resensitized LR HCC cells to lenvatinib treatment. Conclusion: Collectively, our results establish a crucial role of ITGB8 in lenvatinib resistance, and suggest that targeting the ITGB8/HSP90/AKT axis is a promising therapeutic strategy in patients with HCC exhibiting lenvatinib resistance.
- Published
- 2022
- Full Text
- View/download PDF
3. Targeting EphA2 suppresses hepatocellular carcinoma initiation and progression by dual inhibition of JAK1/STAT3 and AKT signaling
- Author
-
Hao Wang, Wei Hou, Aldeb Perera, Carlee Bettler, Jordan R. Beach, Xianzhong Ding, Jun Li, Mitchell F. Denning, Asha Dhanarajan, Scott J. Cotler, Cara Joyce, Jun Yin, Fowsiyo Ahmed, Lewis R. Roberts, and Wei Qiu
- Subjects
EphA2 ,HCC ,AKT ,STAT3 ,JAK1 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Hepatocellular carcinoma (HCC) remains one of the deadliest malignancies worldwide. One major obstacle to treatment is a lack of effective molecular-targeted therapies. In this study, we find that EphA2 expression and signaling are enriched in human HCC and associated with poor prognosis. Loss of EphA2 suppresses the initiation and growth of HCC both in vitro and in vivo. Furthermore, CRISPR/CAS9-mediated EphA2 inhibition significantly delays tumor development in a genetically engineered murine model of HCC. Mechanistically, we discover that targeting EphA2 suppresses both AKT and JAK1/STAT3 signaling, two separate oncogenic pathways in HCC. We also identify a small molecule kinase inhibitor of EphA2 that suppresses tumor progression in a murine HCC model. Together, our results suggest EphA2 as a promising therapeutic target for HCC.
- Published
- 2021
- Full Text
- View/download PDF
4. Systemic Therapy in Metastatic Hepatocellular Carcinoma
- Author
-
Destry Elms, Ami Badami, and Asha Dhanarajan
- Subjects
Carcinoma, Hepatocellular ,Liver Neoplasms ,Gastroenterology ,Humans ,General Medicine ,Immunotherapy ,Sorafenib - Abstract
Multiple new tyrosine kinase inhibitors, immunotherapies and anti-angiogenic therapies are now available for the treatment of advanced hepatocellular carcinoma (HCC). In this article, we reviewed the evidence supporting these new therapies.The combination of atezolizumab and bevacizumab has become a new standard of care for initial systemic therapy in eligible patients, replacing sorafenib in the first line for many patients. Lenvatinib, a multikinase inhibitor, is also a new first line treatment option for patients who are not eligible for immunotherapy. Several additional options for second line treatment were also reviewed in detail in this paper. New systemic therapies for advanced HCC have prolonged overall survival. However, these new therapies are primarily approved for patients with Child-Pugh A classification with few options for patients with Child-Pugh B disease. Further work is needed to expand options for patients with more advanced liver disease and to optimize the sequencing of these new therapies.
- Published
- 2022
5. Focal Adhesion Kinase and β‐Catenin Cooperate to Induce Hepatocellular Carcinoma
- Author
-
Peter Breslin, Hao Wang, Gina Kuffel, Cara Joyce, Na Shang, Aldeb Perera, Thomas Bank, Scott J. Cotler, Asha Dhanarajan, Michael J. Zilliox, Wei Qiu, and Xianzhong Ding
- Subjects
Male ,0301 basic medicine ,Carcinoma, Hepatocellular ,medicine.disease_cause ,Article ,Focal adhesion ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Kinase activity ,beta Catenin ,Regulation of gene expression ,Hepatology ,Chemistry ,Liver Neoplasms ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Androgen receptor ,030104 developmental biology ,Tyrosine kinase 2 ,Focal Adhesion Kinase 1 ,Catenin ,Mutation ,Cancer research ,Female ,030211 gastroenterology & hepatology ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Carcinogenesis - Abstract
There is an urgent need to understand the molecular signaling pathways that drive or mediate the development of hepatocellular carcinoma (HCC). The focal adhesion kinase (FAK) gene, PTK2, is amplified in 16.4% of The Cancer Genome Atlas HCC specimens, and its amplification leads to increased FAK messenger RNA expression. It is not known whether the overexpression of FAK alone is sufficient to induce HCC or whether it must cooperate in some ways with other oncogenes. In this study, we found that 34.8% of human HCC samples with FAK amplification also show β-catenin mutations, suggesting a co-occurrence of FAK overexpression and β-catenin mutations in HCC. We overexpressed FAK alone, constitutively active forms of β-catenin (CAT) alone, or a combination of FAK and CAT in the livers of C57/BL6 mice. We found that overexpression of both FAK and CAT, but neither FAK nor CAT alone, in mouse livers was sufficient to lead to tumorigenesis. We further demonstrated that FAK’s kinase activity is required for FAK/CAT-induced tumorigenesis. Furthermore, we performed RNA sequencing analysis to identify the genes/signaling pathways regulated by FAK, CAT, or FAK/CAT. We found that FAK overexpression dramatically enhances binding of β-catenin to the promoter of androgen receptor (AR), which leads to increased expression of AR in mouse livers. Moreover, ASC-J9, an AR degradation enhancer, suppressed FAK/β-catenin-induced HCC formation. Conclusion: FAK overexpression and β-catenin mutations often co-occur in human HCC tissues. Co-overexpression of FAK and CAT leads to HCC formation in mice through an increased expression of AR. This mouse model will be useful for further studies of the molecular mechanisms in the pathogenesis of HCC and could lead to the identification of new therapeutic targets.
- Published
- 2019
- Full Text
- View/download PDF
6. Combined Use of Aspirin and Statin is Associated With a Decreased Incidence of Hepatocellular Carcinoma
- Author
-
Scott J. Cotler, Benjamin Schmidt, Daniel Aldrich, Steven Scaglione, David Park, Chris Kasia, Asha Dhanarajan, Amy Wozniak, and Jasleen Singh
- Subjects
medicine.medical_specialty ,Statin ,Carcinoma, Hepatocellular ,medicine.drug_class ,Esophageal and Gastric Varices ,Gastroenterology ,Risk Factors ,Internal medicine ,Medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Retrospective Studies ,Aspirin ,business.industry ,Incidence (epidemiology) ,Incidence ,Hazard ratio ,Liver Neoplasms ,Retrospective cohort study ,medicine.disease ,digestive system diseases ,Confidence interval ,Hepatocellular carcinoma ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,business ,Gastrointestinal Hemorrhage ,medicine.drug - Abstract
Background Hepatocellular carcinoma (HCC) is a leading cause of cancer and cancer-related mortality worldwide. Studies have suggested that aspirin (ASA) and statins may be associated with a decrease in incident HCC. Goals We aimed to evaluate the effect of ASA and statin use on the incidence of HCC in a prospective cohort of patients with liver cirrhosis and to identify if there was an increased risk of esophageal variceal hemorrhage (VH) in patients with ASA use. Study We conducted a retrospective study of 521 patients with data collected from July 1, 2012 to December 31, 2017. We used competing risk analysis to assess the association between risk factors and HCC; and the association between ASA and VH. Results ASA use alone was associated with a decreased incidence of HCC in the univariate and multivariate models; [hazard ratio (HR) confidence interval (CI): 0.348 (0.124-0.976); P=0.0448] and [HR (CI): 0.266 (0.094-0.755); P=0.0129, respectively]. The combination of ASA and statin use was associated with a decreased hazard of HCC [HR (CI): 0.15 (0.036-0.624); P=0.0090] and this remained statistically significant in the multivariable model [HR (CI): 0.113 (0.026-0.483); P=0.0033]. Among daily ASA users compared with non-users, there was not a significant increase in risk of VH. Conclusions Daily ASA use was associated with a decrease risk of incident HCC. The combination of daily ASA use and statin use decreased the risk of incident HCC suggesting there is beneficial interaction. Finally, no excess VH was observed in daily ASA users compared with non-users.
- Published
- 2020
7. Advanced esophagogastric cancers: the making or breaking of a backbone
- Author
-
Asha Dhanarajan and Shelly Lo
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2017
- Full Text
- View/download PDF
8. ABL1, Overexpressed in Hepatocellular Carcinomas, Regulates Expression of NOTCH1 and Promotes Development of Liver Tumors in Mice
- Author
-
Anthony J. Koleske, Thomas Bank, Wei Hou, Lennox Chitsike, Fang Wang, Mitchell F. Denning, Scott J. Cotler, Aldeb Perera, Xianzhong Ding, Jun Li, Peter Breslin, Wei Qiu, Wenan Qiang, Carlee Bettler, Yingchen Xu, and Asha Dhanarajan
- Subjects
Male ,0301 basic medicine ,Carcinoma, Hepatocellular ,Datasets as Topic ,Kaplan-Meier Estimate ,Biology ,Proto-Oncogene Mas ,Article ,Proto-Oncogene Proteins c-myc ,Small hairpin RNA ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,hemic and lymphatic diseases ,Gene expression ,microRNA ,Animals ,Humans ,Phosphorylation ,Receptor, Notch1 ,Proto-Oncogene Proteins c-abl ,Gene knockdown ,Hepatology ,Oncogene ,Cell growth ,Liver Neoplasms ,Gastroenterology ,Prognosis ,Xenograft Model Antitumor Assays ,digestive system diseases ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Pyrimidines ,030104 developmental biology ,Liver ,Gene Knockdown Techniques ,Catenin ,Cancer research ,Female ,030211 gastroenterology & hepatology ,Tyrosine kinase - Abstract
BACKGROUND & AIMS: We investigated whether ABL proto-oncogene 1, non-receptor tyrosine kinase (ABL1) is involved in development of hepatocellular carcinoma (HCC). METHODS: We analyzed clinical and gene expression data from The Cancer Genome Atlas. Albumin-Cre (Hep(WT)) mice and mice with hepatocyte-specific disruption of Abl1 (Hep(Abl−/−) mice) were given hydrodynamic injections of plasmids encoding the sleeping beauty transposase and transposons with the MET gene and a catenin beta 1 gene with an N-terminal truncation, which induces development of liver tumors. Some mice were then gavaged with the ABL1 inhibitor nilotinib or vehicle (control), daily for 4 weeks. We knocked down ABL1 with short hairpin RNAs in Hep3B and Huh7 HCC cells and analyzed their proliferation and growth as xenograft tumors in mice. We performed RNA sequencing and gene set enrichment analysis of tumors. We knocked down or overexpressed NOTCH1 and MYC in HCC cells and analyzed proliferation. We measured levels of phosphorylated ABL1, MYC, and NOTCH1 by immunohistochemical analysis of an HCC tissue microarray. RESULTS: HCC tissues had higher levels of ABL1 than non-tumor liver tissues, which correlated with shorter survival times of patients. Hep(WT) mice with the MET and catenin beta 1 transposons developed liver tumors and survived a median 64 days; Hep(Abl−/−) mice with these transposons developed tumors that were 50% smaller and survived a median 81 days. Knockdown of ABL1 in human HCC cells reduced proliferation, growth as xenograft tumors in mice, and expression of MYC, which reduced expression of NOTCH1. . Knockdown of NOTCH1 or MYC in HCC cells significantly reduced cell growth. NOTCH1 or MYC overexpression in human HCC cells promoted proliferation and rescued the phenotype caused by ABL1 knockdown. The level of phosphorylated (activated) ABL1 correlated with levels of MYC and NOTCH1 in human HCC specimens. Nilotinib decreased the expression of MYC and NOTCH1 in HCC cell lines, reduced their growth of xenograft tumors in mice, and slowed growth of liver tumors in mice with MET and catenin beta 1 transposons, reducing tumor levels of MYC and NOTCH1. CONCLUSIONS: HCC samples have increased levels of ABL1 compared with non-tumor liver tissues, and increased levels of ABL1 correlate with shorter survival times of patients. Loss or inhibition of ABL1 reduces proliferation of HCC cells and slows growth of liver tumors in mice. Inhibitors of ABL1 might be used for treatment of HCC.
- Published
- 2020
- Full Text
- View/download PDF
9. Randomized trial of standard chemotherapy alone or combined with atezolizumab as adjuvant therapy for patients with stage III colon cancer and deficient mismatch repair (ATOMIC, Alliance A021502)
- Author
-
Deirdre Jill Cohen, Jeffrey A. Meyerhardt, Kabir Mody, Andrew B. Nixon, Robert J. Behrens, Alexandra Levasseur, Nicolas Sommer, Tyler Zemla, Eileen M. O'Reilly, Amylou C. Dueck, Fang-Shu Ou, Christopher H. Lieu, Frank A. Sinicrope, Asha Dhanarajan, Federico Innocenti, and Walter R. Peters
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,medicine.medical_treatment ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Atezolizumab ,Internal medicine ,medicine ,Adjuvant therapy ,Chemotherapy ,biology ,business.industry ,medicine.disease ,Stage III Colon Cancer ,030220 oncology & carcinogenesis ,biology.protein ,DNA mismatch repair ,Antibody ,business ,030215 immunology - Abstract
e15169 Background: In metastatic colorectal cancer with deficient DNA mismatch repair (dMMR), anti-PD-1 antibody monotherapy produced high tumor response rates and extended progression-free survival compared to lack of benefit for cancers with proficient MMR. In an ongoing phase III randomized trial, we will determine if the addition of the anti-PD-L1 antibody, atezolizumab (Genentech), to adjuvant FOLFOX can improve patient disease-free survival (DFS) vs FOLFOX alone in patients with stage III colon cancers with dMMR. By blocking the PD-1/PD-L1 interaction, atezolizumab may activate T cells, thereby, restoring their ability to detect and attack tumor cells. Limited data suggest that FOLFOX may increase intratumoral cytotoxic CD8+ T cells that could serve as ‘immune priming'. Methods: Patients with curatively resected stage III colon carcinomas with evidence of dMMR are randomized to modified FOLFOX6 for 6 months (12 cycles) alone (control arm) or combined with atezolizumab (840 mg IV q2 wk) with continuation of the antibody as monotherapy for an additional 6 months (total duration of 12 months) [experimental arm]. Patients will be stratified by T, N stage and tumor sidedness. Local testing for MMR proteins is allowed. Atezolizumab must begin by/with cycle 2. One cycle of FOLFOX is allowed pre-registration. The targeted accrual goal of 700 patients and 165 disease-free survival (DFS) events will provide 90% power to detect an effect size expressed as hazard ratio of 0.6 for the primary endpoint of DFS at two-sided alpha of 0.05. Interim analyses are planned at 50% and 75% of events. Secondary endpoints include overall survival, treatment tolerability, and quality of life. Results: This study is being conducted by the Alliance for Clinical Trials in Oncology, was approved by NCI CTEP and activated in 09/2017. The study is actively accruing and, as of 02/11/2019, 152 patients are enrolled. We are actively exploring an international collaboration. Conclusions: This is a current clinical trial in progress. Clinical trial information: NCT02912559.
- Published
- 2019
- Full Text
- View/download PDF
10. Randomized trial of FOLFOX alone or combined with atezolizumab as adjuvant therapy for patients with stage III colon cancer and deficient DNA mismatch repair or microsatellite instability (ATOMIC, Alliance A021502)
- Author
-
Jeffrey A. Meyerhardt, Qian Shi, Kabir Mody, Andrew B. Nixon, Alexandra Levasseur, Frank A. Sinicrope, Walter R. Peters, Amylou C. Dueck, Eileen M. O'Reilly, Deirdre Jill Cohen, Daniel J. Sargent, Federico Innocenti, Christopher H. Lieu, Asha Dhanarajan, Robert J. Behrens, Fang-Shu Ou, and Nicolas Sommer
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,FOLFOX ,Randomized controlled trial ,law ,Atezolizumab ,Internal medicine ,medicine ,Adjuvant therapy ,biology ,business.industry ,Microsatellite instability ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,DNA mismatch repair ,Antibody ,business ,medicine.drug - Abstract
TPS3630 Background: In metastatic colorectal cancer with deficient DNA mismatch repair (MMR), anti-PD-1 antibody monotherapy produced high tumor response rates and extended progression-free survival compared to lack of benefit for proficient MMR tumors (Le, M, et al, NEJM 2016). We propose a phase III randomized trial to determine if the addition of the anti-PD-L1 antibody, atezolizumab (Genentech™), to adjuvant FOLFOX can improve patient disease-free survival (DFS) vs FOLFOX alone in patients with stage III colon cancers with dMMR or microsatellite instability (MSI). By blocking the PD-1/PD-L1 interaction, atezolizumab may activate T cells, thereby, restoring their ability to detect and attack tumor cells. Limited data suggest that FOLFOX may increase intratumoral cytotoxic CD8+ T cells that may serve as ‘immune priming.’ Methods: Patients with curatively resected stage III colon carcinomas with evidence of dMMR or MSI will be randomized to modified FOLFOX6 for 6 months (12 cycles) alone or combined with atezolizumab (840 mg IV q2 wk) continued as monotherapy for an additional 6 months (total duration of 12 months). Patients will be stratified by T, N stage and tumor sidedness. Local testing for MSI or MMR proteins is allowed. Atezolizumab must begin by/with cycle 2. The targeted accrual goal of 700 patients provides 90% power to detect an effect size expressed as hazard ratio of 0.6 for the primary endpoint DFS at two-sided alpha of 0.05. Interim analyses are planned at 50% and 75% of events. Secondary endpoints include overall survival, treatment tolerability, and quality of life. This study will be conducted by the Alliance for Clinical Trials in Oncology. The protocol has been approved by NCI CTEP and is expected to be activated in mid 2017. Clinical trial information: NCT02912559.
- Published
- 2017
- Full Text
- View/download PDF
11. Expression of GRP78 protein in pancreatic ductal adenocarcinoma and intraductal papillary mucinous neoplasm
- Author
-
Gerard Abood, Jennifer L. Gnerlich, Xianzhong Ding, Sam G. Pappas, Asha Dhanarajan, and Reginald Hill
- Subjects
Cancer Research ,Chemotherapy ,Pathology ,medicine.medical_specialty ,endocrine system diseases ,Intraductal papillary mucinous neoplasm ,business.industry ,Endoplasmic reticulum ,medicine.medical_treatment ,medicine.disease ,Staining ,Exact test ,Oncology ,Cancer cell ,medicine ,Immunohistochemistry ,business ,Immunostaining - Abstract
323 Background: Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis due to late detection, aggressive biology and resistance to chemotherapy. Markers for early detection of PDAC and mediators of chemoresistance are poorly understood. GRP78 is a major endoplasmic reticulum (ER) chaperone protein critical for protein quality control of the ER. Increased GRP78 expression promotes in vitro cancer cell survival, progression, and chemoresistance. This study aims to examine expression of GRP78 protein in PDAC and intraductal papillary mucinous neoplasm (IPMN) compared to normal pancreatic ducts using immunohistochemistry (IHC). Methods: The expression of GRP78 was assessed using IHC in 93 sections of formalin-fixed paraffin-embedded tissue: normal ducts (38), low grade IPMN (IPMN-LG, 26), high grade IPMN (IPMN-HG, 9), and PDAC (20). Immunostaining intensity of GRP78 protein was categorized as no or weak staining (0-1+) and strong staining (2-3+). Fisher’s exact test with two tails was performed using the GraphPad statistical software. Results: GRP78 expression was identified in the cytoplasm of normal pancreatic ducts, IPMN and PDAC with a fine granular pattern. All PDAC and IPMN-HG showed strong expression of GRP78 while the normal ductal cells showed only minimal expression of GRP78. 73% of IPMN-LG expresses GRP78 strongly. Statistical analysis revealed a significant difference in GRP78 expression level between normal ductal cells and all three pathological conditions including PDCA, IPMN-HG and IPMN-LG (all p < 0.0001). Conclusions: This study shows that the expression level of GRP78 is significantly increased in PDAC, IPMN-HG and IPMN-LG compared to normal ductal cells. There appears to be a progressive increase in GRP78 expression from IPMN-LG to IPMN-HG and invasive PDAC. Increased GRP78 expression may be a marker for those at risk of developing PDAC. Changes in GRP78 expression over time and its role in PDAC chemoresistance should be further assessed. [Table: see text]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.