412 results on '"Dasari, Arvind"'
Search Results
402. Progression-free survival remains poor over sequential lines of systemic therapy in patients with BRAF-mutated colorectal cancer.
- Author
-
Morris V, Overman MJ, Jiang ZQ, Garrett C, Agarwal S, Eng C, Kee B, Fogelman D, Dasari A, Wolff R, Maru D, and Kopetz S
- Subjects
- Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized administration & dosage, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Cetuximab, Colorectal Neoplasms pathology, Disease-Free Survival, Female, Humans, Irinotecan, Liver Neoplasms secondary, Liver Neoplasms surgery, Lung Neoplasms secondary, Lymphatic Metastasis, Male, Metastasectomy, Microsatellite Instability, Middle Aged, Mutation, Neoplasm Staging, Organoplatinum Compounds administration & dosage, Oxaliplatin, Panitumumab, Peritoneal Neoplasms secondary, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Liver Neoplasms drug therapy, Lung Neoplasms drug therapy, Peritoneal Neoplasms drug therapy, Proto-Oncogene Proteins B-raf genetics
- Abstract
Background: BRAF mutations occur in 5% to 10% of metastatic colorectal cancers and are biomarkers associated with a poor prognosis. However, the outcomes with standard chemotherapy over sequential lines of therapy in a large cohort of patients with BRAF-mutant tumors have not been described., Patients and Methods: We searched the M.D. Anderson Cancer Center databases for patients with colorectal cancer and identified BRAF mutations between December 2003 and May 2012. Patients were analyzed for clinical characteristics, PFS, overall survival, and chemotherapeutic agents used. Survival was estimated according to the Kaplan-Meier method., Results: Among the 1567 patients tested for BRAF mutations at our institution, 127 (8.1%) had tumors with BRAF mutations. The 71 patients who presented with metastatic disease received a median of 2 lines of chemotherapy. For the first 3 lines of chemotherapy, median PFS was 6.3 months (n = 69 patients; 95% confidence interval [CI], 4.9-7.7 months), 2.5 months (n = 58 patients; 95% CI, 1.8-3.0 months), and 2.6 months (n = 31 patients; 95% CI, 1.0-4.2 months), respectively. Median PFS was not affected by the backbone chemotherapeutic agent in the first-line setting, whether oxaliplatin-based or irinotecan-based (6.4 months vs. 5.4 months, respectively; P = .99)., Conclusion: PFS is expectedly poor for patients with BRAF-mutated metastatic colorectal cancer. Despite the ascertainment bias present (with testing preferentially performed in patients suitable for clinical trials in refractory disease), these data provide historic controls suitable for future study design and support the idea that novel therapeutic options are essential in this population., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
403. A phase II study of the gamma secretase inhibitor RO4929097 in patients with previously treated metastatic pancreatic adenocarcinoma.
- Author
-
De Jesus-Acosta A, Laheru D, Maitra A, Arcaroli J, Rudek MA, Dasari A, Blatchford PJ, Quackenbush K, and Messersmith W
- Subjects
- Adenocarcinoma metabolism, Aged, Aged, 80 and over, Amyloid Precursor Protein Secretases metabolism, Antineoplastic Agents pharmacokinetics, Benzazepines pharmacokinetics, Disease-Free Survival, Female, Humans, Male, Middle Aged, Pancreatic Neoplasms metabolism, Survival Rate, Pancreatic Neoplasms, Adenocarcinoma drug therapy, Amyloid Precursor Protein Secretases antagonists & inhibitors, Antineoplastic Agents therapeutic use, Benzazepines therapeutic use, Enzyme Inhibitors therapeutic use, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: The notch pathway is overexpressed in pancreatic adenocarcinoma. RO4929097, an oral inhibitor of the γ-secretase enzyme has been safely given as a single agent in patients with advanced solid tumors. We aimed to evaluate the efficacy of RO4929097 in patients with pancreatic adenocarcinoma (PDA)., Methods: A two-stage, single-arm Phase II trial was conducted in patients with previously treated metastatic PDA. RO4929097 was administered at a dose of 20 mg daily on days 1-3, 8-10 and 15-17 of 21-day cycles. The primary endpoint was survival at 6 months. Secondary endpoints included overall survival (OS), response rate, toxicities, pharmacokinetic and pharmacodynamic analyses., Results: Eighteen patients were recruited, 17 in the first stage and one in the 2nd stage. It was decided to stop further enrollment after RO4929097 was discontinued by the sponsor and was no longer a development candidate. Three (25 %) of 12 evaluable patients achieved stable disease. The 6-month survival rate was 27.8 % (95 % CI 9.7-53.5). The median OS was 4.1 months (95 % CI 2.7-5.8 months) and median progression-free survival was 1.5 months (95 % CI 1.3-1.6 months). Pharmacokinetic properties of RO4929097 in patients (n = 5) with PDA was similar to that previously reported in other patient populations. There was a trend towards a decrease in HeyL (p = 0.08) gene expression in three patients following study drug administration., Conclusions: RO4929097 was well-tolerated in patients with previously treated PDA. Development of RO4929097 has been discontinued, but development of other notch-targeting agents in pancreatic cancer is continuing.
- Published
- 2014
- Full Text
- View/download PDF
404. Atypical metastatic presentations in colorectal cancer: a case series.
- Author
-
Rogers JE, Ohinata A, Dasari A, and Eng C
- Subjects
- Adult, Aged, Female, Humans, Male, Mandibular Neoplasms secondary, Pancreatic Neoplasms secondary, Scalp pathology, Skin Neoplasms secondary, Soft Tissue Neoplasms secondary, Thyroid Neoplasms secondary, Adenocarcinoma secondary, Colorectal Neoplasms pathology
- Published
- 2014
- Full Text
- View/download PDF
405. Phase I pharmacokinetic and pharmacodynamic study of cetuximab, irinotecan and sorafenib in advanced colorectal cancer.
- Author
-
Azad N, Dasari A, Arcaroli J, Taylor GE, Laheru DA, Carducci MA, McManus M, Quackenbush K, Wright JJ, Hidalgo M, Diaz LA Jr, Donehower RC, Zhao M, Rudek MA, and Messersmith WA
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Cetuximab, Dose-Response Relationship, Drug, Female, Humans, Irinotecan, Male, Maximum Tolerated Dose, Middle Aged, Niacinamide administration & dosage, Niacinamide analogs & derivatives, Phenylurea Compounds administration & dosage, Prognosis, Sorafenib, Tissue Distribution, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy
- Abstract
Background This phase Ib study was designed to determine the maximum tolerated doses (MTD) and dose limiting toxicities (DLTs) of irinotecan and cetuximab with sorafenib. Secondary objectives included characterizing the pharmacokinetics and pharmacodynamics and evaluating preliminary antitumor activity in patients with advanced colorectal cancer (CRC). Methods Patients with metastatic, pretreated CRC were treated at five dose levels. Results Eighteen patients were recruited with median age 56.5 years. In the first five patients treated, 2 irinotecan related DLTs were observed. With reduced dose intensity irinotecan, there were no further DLTs. The most common toxicities were diarrhea, nausea/vomiting, fatigue, anorexia and rash. DLTs included neutropenia and thrombocytopenia. Two patients had partial responses (one with a KRAS mutation) and 8 had stable disease (8-36 weeks). The median progression free survival (PFS) and overall survival (OS) were 2.5 and 4.7 months respectively. Pharmacokinetic analyses suggest sorafenib and metabolite exposure correlate with OS and DLTs. Conclusions The recommended phase II dose (RP2D) is irinotecan 100 mg/m(2) i.v. days 1, 8; cetuximab 400 mg/m(2) i.v. days 1 and 250 mg/m(2) i.v. weekly; and sorafenib 400 mg orally twice daily in advanced, pretreated CRC. The combination resulted in a modest response rate.
- Published
- 2013
- Full Text
- View/download PDF
406. Biomarker-driven trial in metastatic pancreas cancer: feasibility in a multicenter study of saracatinib, an oral Src inhibitor, in previously treated pancreatic cancer.
- Author
-
Arcaroli J, Quackenbush K, Dasari A, Powell R, McManus M, Tan AC, Foster NR, Picus J, Wright J, Nallapareddy S, Erlichman C, Hidalgo M, and Messersmith WA
- Subjects
- 3' Untranslated Regions genetics, Administration, Oral, Adult, Aged, Antineoplastic Agents adverse effects, Benzodioxoles adverse effects, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Class I Phosphatidylinositol 3-Kinases, Female, Humans, Insulin-Like Growth Factor Binding Protein 2 genetics, Insulin-Like Growth Factor Binding Protein 2 metabolism, Male, Middle Aged, Neoplasm Metastasis, Pancreatic Neoplasms mortality, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Quinazolines adverse effects, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Antineoplastic Agents administration & dosage, Benzodioxoles administration & dosage, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Quinazolines administration & dosage, src-Family Kinases antagonists & inhibitors
- Abstract
Src tyrosine kinases are overexpressed in pancreatic cancers, and the oral Src inhibitor saracatinib has shown antitumor activity in preclinical models of pancreas cancer. We performed a CTEP-sponsored Phase II clinical trial of saracatinib in previously treated pancreas cancer patients, with a primary endpoint of 6-month survival. A Simon MinMax two-stage phase II design was used. Saracatinib (175 mg/day) was administered orally continuously in 28-day cycles. In the unselected portion of the study, 18 patients were evaluable. Only two (11%) patients survived for at least 6 months, and three 6-month survivors were required to move to second stage of study as originally designed. The study was amended as a biomarker-driven trial (leucine rich repeat containing protein 19 [LRRC19] > insulin-like growth factor-binding protein 2 [IGFBP2] "top scoring pairs" polymerase chain reaction [PCR] assay, and PIK3CA mutant) based on preclinical data in a human pancreas tumor explant model. In the biomarker study, archival tumor tissue or fresh tumor biopsies were tested. Biomarker-positive patients were eligible for the study. Only one patient was PIK3CA mutant in a 3' untranslated region (UTR) portion of the gene. This patient was enrolled in the study and failed to meet the 6-month survival endpoint. As the frequency of biomarker-positive patients was very low (<3%), the study was closed. Although we were unable to conclude whether enriching for a subset of second/third line pancreatic cancer patients treated with a Src inhibitor based on a biomarker would improve 6-month survival, we demonstrate that testing pancreatic tumor samples for a biomarker-driven, multicenter study in metastatic pancreas cancer is feasible.
- Published
- 2012
- Full Text
- View/download PDF
407. Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors.
- Author
-
Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, Gibbon DG, Patnaik A, Falchook GS, Dasari A, Shapiro GI, Boylan JF, Xu ZX, Wang K, Koehler A, Song J, Middleton SA, Deutsch J, Demario M, Kurzrock R, and Wheler JJ
- Subjects
- Adult, Aged, Aged, 80 and over, Benzazepines adverse effects, Benzazepines pharmacokinetics, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Humans, Male, Middle Aged, Neoplasms enzymology, Neoplasms metabolism, Neoplasms pathology, Receptors, Notch metabolism, Signal Transduction drug effects, Amyloid Precursor Protein Secretases antagonists & inhibitors, Benzazepines therapeutic use, Neoplasms drug therapy, Receptors, Notch antagonists & inhibitors
- Abstract
Purpose: To determine the maximum-tolerated dose (MTD) and assess safety, pharmacokinetics, pharmacodynamics, and evidence of antitumor activity of RO4929097, a gamma secretase inhibitor of Notch signaling in patients with advanced solid malignancies., Patients and Methods: Patients received escalating doses of RO4929097 orally on two schedules: (A) 3 consecutive days per week for 2 weeks every 3 weeks; (B) 7 consecutive days every 3 weeks. To assess reversible CYP3A4 autoinduction, the expanded part of the study tested three dosing schedules: (B) as above; modified A, 3 consecutive d/wk for 3 weeks; and (C) continuous daily dosing. Positron emission tomography scans with [(18)F]fluorodeoxyglucose (FDG-PET) were used to assess tumor metabolic effects., Results: Patients on schedule A (n = 58), B (n = 47), and C (n = 5; expanded cohort) received 302 cycles of RO4929097. Common grade 1 to 2 toxicities were fatigue, thrombocytopenia, fever, rash, chills, and anorexia. Transient grade 3 hypophosphatemia (dose-limiting toxicity, one patient) and grade 3 pruritus (two patients) were observed at 27 mg and 60 mg, respectively; transient grade 3 asthenia was observed on schedule A at 80 mg (one patient). Tumor responses included one partial response in a patient with colorectal adenocarcinoma with neuroendocrine features, one mixed response (stable disease) in a patient with sarcoma, and one nearly complete FDG-PET response in a patient with melanoma. Effect on CYP3A4 induction was observed., Conclusion: RO4929097 was well tolerated at 270 mg on schedule A and at 135 mg on schedule B; the safety of schedule C has not been fully evaluated. Further studies are warranted on the basis of a favorable safety profile and preliminary evidence of clinical antitumor activity.
- Published
- 2012
- Full Text
- View/download PDF
408. ALDH+ tumor-initiating cells exhibiting gain in NOTCH1 gene copy number have enhanced regrowth sensitivity to a γ-secretase inhibitor and irinotecan in colorectal cancer.
- Author
-
Arcaroli JJ, Powell RW, Varella-Garcia M, McManus M, Tan AC, Quackenbush KS, Pitts TM, Gao D, Spreafico A, Dasari A, Touban BM, and Messersmith WA
- Subjects
- Aldehyde Dehydrogenase genetics, Amyloid Precursor Protein Secretases antagonists & inhibitors, Animals, Camptothecin pharmacology, Camptothecin therapeutic use, Colorectal Neoplasms genetics, Female, Gene Dosage genetics, Humans, In Situ Hybridization, Fluorescence, Irinotecan, Mice, Mice, Nude, Receptor, Notch1 genetics, Signal Transduction, Xenograft Model Antitumor Assays, Aldehyde Dehydrogenase metabolism, Camptothecin analogs & derivatives, Colorectal Neoplasms drug therapy, Colorectal Neoplasms metabolism, Receptor, Notch1 metabolism
- Abstract
The Notch signaling pathway has been shown to be upregulated in colorectal cancer (CRC) and important for the self-renewal of cancer stem cells. In this study, we evaluated the efficacy of PF-03084014, a γ-secretase inhibitor, in combination with irinotecan to identify the effects of treatment on tumor recurrence and the tumor-initiating population in our CRC preclinical explant model. The combination of PF-03084014 and irinotecan had the greatest effect at reducing tumor growth on four CRC tumors when compared with treatment with PF-03084014 or irinotecan alone. The combination significantly reduced tumor recurrence in two CRC explants (CRC001 and CRC036) after treatment was discontinued. Both of these tumors exhibited elevated baseline levels of Notch pathway activation as well as an increase in NOTCH1 gene copy number when compared with the two CRC explants (CRC026 and CRC027) where tumors reappeared quickly after termination of treatment. Isolation and injection of aldehyde dehydrogenase (ALDH(+) and ALDH(-)) cells in an in vivo explant model demonstrated that the ALDH(+) cell population were tumorigenic. Evaluation of the ALDH(+) cells after 28 days of treatment showed that the combination reduced the ALDH(+) population in the tumors that did not regrow. Furthermore, ALDH(+) cells from CRC001 and CRC027 were injected in vivo and treated immediately for 28 days. Two months after treatment, tumors were evident in the combination treatment group for CRC027 but not for CRC036. These results indicate the combination of PF-03084014 and irinotecan may be effective in reducing tumor recurrence in CRC patients whose tumors exhibit elevated levels of the Notch pathway., (Copyright © 2012 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
409. Common PIK3CA mutants and a novel 3' UTR mutation are associated with increased sensitivity to saracatinib.
- Author
-
Arcaroli JJ, Quackenbush KS, Powell RW, Pitts TM, Spreafico A, Varella-Garcia M, Bemis L, Tan AC, Reinemann JM, Touban BM, Dasari A, Eckhardt SG, and Messersmith WA
- Subjects
- Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases, Colorectal Neoplasms pathology, Gene Dosage, Gene Expression Profiling, Humans, Immunoblotting, Immunoprecipitation, Mice, MicroRNAs genetics, Oligonucleotide Array Sequence Analysis, PTEN Phosphohydrolase, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, src-Family Kinases antagonists & inhibitors, src-Family Kinases genetics, src-Family Kinases metabolism, 3' Untranslated Regions genetics, Benzodioxoles pharmacology, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Drug Resistance, Neoplasm genetics, Mutation genetics, Phosphatidylinositol 3-Kinases genetics, Quinazolines pharmacology
- Abstract
Purpose: Dysregulation of the phosphoinositide 3-kinase (PI3K) and Src signaling pathways commonly occur in colorectal cancer. Mutations in the PIK3CA gene are associated with an increase in severity of disease and worse clinical outcomes. Elevated levels of Src have been identified in premalignant lesions and are suggested to play a central role in tumor progression. Because these pathways appear to enhance tumor growth and metastasis, molecularly targeted agents for both pathways are currently being evaluated in early-phase clinical trials., Experimental Design: We used colorectal cancer cell lines and a patient-derived explant model to investigate the efficacy of saracatinib. Mutations in the PIK3CA were evaluated to examine the association between mutations in the PIK3CA gene and sensitivity to saracatinib., Results: We have identified a subset of patients with a PIK3CA (exon 9 and 20) mutation with increased sensitivity to saracatinib. A novel 3' untranslated region (UTR) mutation was also shown to be associated with increased sensitivity to saracatinib and have a reduced affinity for miR-520a and miR-525a. Importantly, we show that Src inhibition reduces the interaction between Src and p85, subsequently decreasing Akt-dependent signaling., Conclusion: These results indicate that a personalized approach in targeting Src in PIK3CA-mutant patients with colorectal cancers may prove effective in a subset of patients with this genetic alteration., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
410. Recurrent pancreatic adenocarcinoma after pancreatic resection.
- Author
-
Dasari A, McCarter M, McManus MC, Russ P, and Messersmith WA
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma pathology, CA-19-9 Antigen blood, Female, Humans, Middle Aged, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Tomography, X-Ray Computed, Adenocarcinoma surgery, Liver Neoplasms secondary, Pancreatic Neoplasms surgery, Pancreaticoduodenectomy
- Abstract
The patient is a 57-year old Caucasian female who presented with right upper quadrant pain and obstructive jaundice and was diagnosed with resectable pancreatic cancer. She underwent pancreaticoduodenectomy (PD) after preoperative biliary stenting. She subsequently presented to the clinic, where it was noticed that she had an elevated CA 19-9. CT C/A/P revealed multiple new liver lesions.
- Published
- 2010
411. New strategies in colorectal cancer: biomarkers of response to epidermal growth factor receptor monoclonal antibodies and potential therapeutic targets in phosphoinositide 3-kinase and mitogen-activated protein kinase pathways.
- Author
-
Dasari A and Messersmith WA
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Biomarkers, Tumor genetics, Clinical Trials as Topic, Colorectal Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Humans, Biomarkers, Tumor analysis, Colorectal Neoplasms genetics, Drug Resistance, Neoplasm genetics, ErbB Receptors immunology, Mitogen-Activated Protein Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction physiology
- Abstract
Initial experience with the epidermal growth factor receptor monoclonal antibodies (EGFR MoAb) in unselected patients with metastatic colorectal cancer (mCRC) showed that most of the treated patients did not derive therapeutic benefit. This outcome has driven the search for biomarkers for this population. Recent advances have further shown the heterogeneous nature of this disease with multiple interlinked pathways being implicated. Two such pathways downstream to the EGFR, mitogen-activated protein kinase (MAPK) and (phosphoinositide 3-kinase) PI3K, have gained increasing attention and become targets for development of novel biomarkers and therapeutic agents. Here, we highlight recent progress., ((c) 2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
412. Oxidative stress induces premature senescence by stimulating caveolin-1 gene transcription through p38 mitogen-activated protein kinase/Sp1-mediated activation of two GC-rich promoter elements.
- Author
-
Dasari A, Bartholomew JN, Volonte D, and Galbiati F
- Subjects
- Animals, Antioxidants pharmacology, Caveolin 1 genetics, Caveolin 1 metabolism, Cell Line, Tumor, Cellular Senescence drug effects, Cellular Senescence physiology, Enzyme Activation, Humans, Hydrogen Peroxide pharmacology, Mice, NIH 3T3 Cells, Oxidants pharmacology, Oxidative Stress genetics, Oxidative Stress physiology, Promoter Regions, Genetic, Quercetin pharmacology, Response Elements, Sp1 Transcription Factor biosynthesis, Sp1 Transcription Factor genetics, Transcription, Genetic, Up-Regulation, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Caveolin 1 biosynthesis, Sp1 Transcription Factor metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Cellular senescence is believed to represent a natural tumor suppressor mechanism. We have previously shown that up-regulation of caveolin-1 was required for oxidative stress-induced premature senescence in fibroblasts. However, the molecular mechanisms underlying caveolin-1 up-regulation in senescent cells remain unknown. Here, we show that subcytotoxic oxidative stress generated by hydrogen peroxide application promotes premature senescence and stimulates the activity of a (-1,296) caveolin-1 promoter reporter gene construct in fibroblasts. Functional deletion analysis mapped the oxidative stress response elements of the mouse caveolin-1 promoter to the sequences -244/-222 and -124/-101. The hydrogen peroxide-mediated activation of both Cav-1 (-244/-222) and Cav-1 (-124/-101) was prevented by the antioxidant quercetin. Combination of electrophoretic mobility shift studies, chromatin immunoprecipitation analysis, Sp1 overexpression experiments, as well as promoter mutagenesis identifies enhanced Sp1 binding to two GC-boxes at -238/-231 and -118/-106 as the core mechanism of oxidative stress-triggered caveolin-1 transactivation. In addition, signaling studies show p38 mitogen-activated protein kinase (MAPK) as the upstream regulator of Sp1-mediated activation of the caveolin-1 promoter following oxidative stress. Inhibition of p38 MAPK prevents the oxidant-induced Sp1-mediated up-regulation of caveolin-1 protein expression and development of premature senescence. Finally, we show that oxidative stress induces p38-mediated up-regulation of caveolin-1 and premature senescence in normal human mammary epithelial cells but not in MCF-7 breast cancer cells, which do not express caveolin-1 and undergo apoptosis. This study delineates for the first time the molecular mechanisms that modulate caveolin-1 gene transcription upon oxidative stress and brings new insights into the redox control of cellular senescence in both normal and cancer cells.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.