11 results on '"Villalona-Calero, Miguel"'
Search Results
2. Ablative Radiotherapy as a Strategy to Overcome TKI Resistance in EGFR-Mutated NSCLC.
- Author
-
Novak, Jennifer, Salgia, Ravi, West, Howard, Villalona-Calero, Miguel A, Sampath, Sagus, Williams, Terence, Villaflor, Victoria, Massarelli, Erminia, Pathak, Ranjan, Koczywas, Marianna, Chau, Brittney, and Amini, Arya
- Subjects
LUNG cancer ,GENETIC mutation ,EPIDERMAL growth factor receptors ,METASTASIS ,PROTEIN-tyrosine kinase inhibitors ,RADIOSURGERY ,DRUG resistance in cancer cells - Abstract
Simple Summary: Most patients with EGFR-mutated NSCLC who receive treatment with targeted therapy will eventually develop resistance, meaning the therapy will lose its efficacy. Prior studies have shown a benefit to continuing to treat patients on TKI therapy despite limited progression of one or more sites of metastatic disease in EGFR-mutated NSCLC. Based on the data reviewed here, the use of radiation therapy to sites of disease progression is both efficacious and carries a low risk for side effects, with the added benefit of allowing patients to continue on TKI therapy. Tyrosine kinase inhibitor (TKI) therapy is the recommended first-line treatment for metastatic non-small-cell lung cancer (NSCLC) positive for epidermal growth factor receptor (EGFR) gene mutation. However, most individuals treated with TKI therapy for EGFR-mutant NSCLC will develop tumor resistance to TKI therapy. Therapeutic strategies to overcome TKI resistance are the topic of several ongoing clinical trials. One potential strategy, which has been explored in numerous trials, is the treatment of progressive sites of disease with stereotactic body radiation treatment (SBRT) or stereotactic radiosurgery (SRS). We sought to review the literature pertaining to the use of local ablative radiation therapy in the setting of acquired resistance to TKI therapy and to discuss stereotactic radiation therapy as a strategy to overcome TKI resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
3. MiRNA-200C expression in Fanconi anemia pathway functionally deficient lung cancers.
- Author
-
Duan, Wenrui, Tang, Shirley, Gao, Li, Dotts, Kathleen, Fink, Andrew, Kalvala, Arjun, Aguila, Brittany, Wang, Qi-En, and Villalona-Calero, Miguel A.
- Subjects
MICRORNA ,GENE expression ,LUNG cancer ,FANCONI'S anemia ,DNA damage - Abstract
The Fanconi Anemia (FA) pathway is essential for human cells to maintain genomic integrity following DNA damage. This pathway is involved in repairing damaged DNA through homologous recombination. Cancers with a defective FA pathway are expected to be more sensitive to cross-link based therapy or PARP inhibitors. To evaluate downstream effectors of the FA pathway, we studied the expression of 734 different micro RNAs (miRNA) using NanoString nCounter miRNA array in two FA defective lung cancer cells and matched control cells, along with two lung tumors and matched non-tumor tissue samples that were deficient in the FA pathway. Selected miRNA expression was validated with real-time PCR analysis. Among 734 different miRNAs, a cluster of microRNAs were found to be up-regulated including an important cancer related micro RNA, miR-200C. MiRNA-200C has been reported as a negative regulator of epithelial-mesenchymal transition (EMT) and inhibits cell migration and invasion by promoting the upregulation of E-cadherin through targeting ZEB1 and ZEB2 transcription factors. miRNA-200C was increased in the FA defective lung cancers as compared to controls. AmpliSeq analysis showed significant reduction in ZEB1 and ZEB2 mRNA expression. Our findings indicate the miRNA-200C potentially play a very important role in FA pathway downstream regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
4. Type of TP53 mutation influences oncogenic potential and spectrum of associated K‐ras mutations in lung‐specific transgenic mice.
- Author
-
Duan, Wenrui, Gao, Li, Kalvala, Arjun, Aguila, Brittany, Brooks, Christopher, Mo, Xiaokui, Ding, Haiming, Shilo, Konstantin, Otterson, Gregory A., and Villalona‐Calero, Miguel A.
- Subjects
TRANSGENIC mice ,NON-small-cell lung carcinoma ,GENETIC code ,LUNG tumors ,LUNG cancer - Abstract
TP53 and K‐ras mutations are two of the major genetic alterations in human nonsmall cell lung cancers. The association between these two genes during lung tumorigenesis is unknown. We evaluated the potential of two common Type I (273H, contact) and Type II (175H, conformational) TP53 mutations to induce lung tumors in transgenic mice, as well as K‐ras status, and other driver mutations in these tumors. Among 516 (138 nontransgenic, 207 SPC‐TP53‐273H, 171 SPC‐TP53‐175H) mice analyzed, 91 tumors, all adenocarcinomas, were observed. Type II mutants developed tumors more frequently (as compared to nontransgenics, p = 0.0003; and Type I, p = 0.010), and had an earlier tumor onset compared to Type I (p = 0.012). K‐ras mutations occurred in 21 of 50 (42%) of murine lung tumors sequenced. For both the nontransgenic and the SPC‐TP53‐273H transgenics, tumor K‐ras codon 12–13 mutations occurred after 13 months with a peak incidence at 16–18 months. However, for the SPC‐TP53‐175H transgenics, K‐ras codon 12–13 mutations were observed as early as 6 months, with a peak incidence between the ages of 10–12 months. Codons 12–13 transversion mutations were the predominant changes in the SPC‐TP53‐175H transgenics, whereas codon 61 transition mutations were more common in the SPC‐TP53‐273H transgenics. The observation of accelerated tumor onset, early appearance and high frequency of K‐ras codon 12–13 mutations in the Type II TP53‐175H mice suggests an enhanced oncogenic function of conformational TP53 mutations, and gains in early genetic instability for tumors containing these mutations compared to contact mutations. What's new? Mutations in TP53 and K‐ras frequently occur in non‐small cell lung cancer. Interactions between these two genes during lung tumorigenesis, however, are unknown. Here, contact‐(273H) and conformational‐(175H) mutations in TP53 were investigated for their role in lung tumorigenesis and for associations with K‐ras mutations. In TP53‐273H mice, transition mutations in K‐ras codon 61 dominated. Meanwhile, in TP53‐175H transgenic mice, transversion mutations in codons 12‐13 were the primary changes. K‐ras codon 12‐13 mutations occurred at a relatively early age in TP53‐175H mice, which also experienced accelerated lung cancer onset. The findings suggest that conformational TP53 mutations possess enhanced tumorigenic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
5. Oncolytic reovirus in combination with chemotherapy in metastatic or recurrent non-small cell lung cancer patients with KRAS-activated tumors.
- Author
-
Villalona‐Calero, Miguel A., Lam, Elaine, Otterson, Gregory A., Zhao, Weiqiang, Timmons, Matthew, Subramaniam, Deepa, Hade, Erinn M., Gill, George M., Coffey, Matthew, Selvaggi, Giovanni, Bertino, Erin, Chao, Bo, and Knopp, Michael V.
- Subjects
- *
TUMORS , *EPIDERMAL growth factor , *NON-small-cell lung carcinoma , *GENETIC mutation , *IMMUNE response , *ANTINEOPLASTIC agents , *BIOTHERAPY , *CANCER relapse , *LUNG cancer , *LUNG tumors , *PROGNOSIS , *PROTEINS , *RESEARCH funding , *RNA viruses , *KAPLAN-Meier estimator , *TUMOR grading - Abstract
Background: The type 3 Dearing reovirus (Reolysin) is a naturally occurring virus that preferentially infects and causes oncolysis in tumor cells with a Ras-activated pathway. It induces host immunity and cell cycle arrest and acts synergistically with cytotoxic agents.Methods: This study evaluated Reolysin combined with paclitaxel and carboplatin in patients with metastatic/recurrent KRAS-mutated or epidermal growth factor receptor (EGFR)-mutated/amplified non-small cell lung cancer.Results: Thirty-seven patients were treated. Molecular alterations included 20 KRAS mutations, 10 EGFR amplifications, 3 EGFR mutations, and 4 BRAF-V600E mutations. In total, 242 cycles (median, 4; range, 1-47) were completed. The initial doses were area under the curve (AUC) 6 mg/mL/min for carboplatin, 200 mg/m(2) for paclitaxel on day 1, and 3 × 10(10) 50% tissue culture infective dose for Reolysin on days 1 to 5 of each 21-day cycle. Because of diarrhea and febrile neutropenia (in the first 2 patients), subsequent doses were reduced to 175 mg/m(2) for paclitaxel and AUC 5 mg/mL/min for carboplatin. Toxicities included fatigue, diarrhea, nausea/vomiting, neutropenia, arthralgia/myalgia, anorexia, and electrolyte abnormalities. Response Evaluation Criteria in Solid Tumors 1.0 responses included the following: partial response for 11 patients, stable disease (SD) for 20 patients, progressive disease for 4 patients, and not evaluable for 2 patients (objective response rate, 31%; 90% 1-sided lower confidence interval, 21%). Four SD patients had >40% positron emission tomography standardized uptake value reductions. The median progression-free survival, median overall survival, and 12-month overall survival rate were 4 months, 13.1 months, and 57%, respectively. Seven patients were alive after a median follow-up of 34.2 months; they included 2 patients without disease progression at 37 and 50 months.Conclusions: Reolysin in combination with paclitaxel and carboplatin was well tolerated. The observed response rate suggests a benefit of the reovirus for chemotherapy. A follow-up randomized study is recommended. The proportion of patients surviving longer than 2 years (30%) suggests a second/third-line treatment effect or possibly the triggering of an immune response after tumor reovirus infiltration. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
6. Fanconi anemia repair pathway dysfunction, a potential therapeutic target in lung cancer.
- Author
-
Li Gao, Aguila, Brittany, Kalvala, Arjun, Duan, Wenrui, Otterson, Gregory A., and Villalona-Calero, Miguel A.
- Subjects
LUNG cancer treatment ,FANCONI'S anemia ,CELL survival ,DNA repair ,WESTERN immunoblotting - Abstract
The Fanconi anemia (FA) pathway is a major mechanism of homologous recombination DNA repair. The functional readout of the pathway is activation through mono-ubiquitination of FANCD2 leading to nuclear foci of repair. We have recently developed an FA triple-staining immunofluorescence based method (FATSI) to evaluate FANCD2 foci formation in formalin fixed paraffin-embedded (FFPE) tumor samples. DNA-repair deficiencies have been considered of interest in lung cancer prevention, given the persistence of damage produced by cigarette smoke in this setting, as well as in treatment, given potential increased efficacy of DNA-damaging drugs. We screened 139 non-small cell lung cancer (NSCLC) FFPE tumors for FANCD2 foci formation by FATSI analysis. Among 104 evaluable tumors, 23 (22%) were FANCD2 foci negative, thus repair deficient. To evaluate and compare novel-targeted agents in the background of FA deficiency, we utilized RNAi technology to render several lung cancer cell lines FANCD2 deficient. Successful FANCD2 knockdown was confirmed by reduction in the FANCD2 protein. Subsequently, we treated the FA defective H1299D2- down and A549D2-down NSCLC cells and their FA competent counterparts (empty vector controls) with the PARP inhibitors veliparib (ABT-888) (5mM) and BMN673 (0.5mM), as well as the CHK1 inhibitor Arry-575 at a dose of 0.5mM.We also treated the FA defective small cell lung cancer cell lines H719D2-down and H792D2-down and their controls with the BCL-2/XL inhibitor ABT-263 at a dose of 2mM. The treated cells were harvested at 24, 48, and 72 h post treatment. MTT cell viability analysis showed that each agent was more cytotoxic to the FANCD2 knock-down cells. In all tests, the FA defective lung cancer cells had less viable cells as comparing to controls 72 h post treatment. Both MTT and clonogenic analyses comparing the two PARP inhibitors, showed that BMN673 was more potent compared to veliparib. Given that FA pathway plays essential roles in response to DNA damage, our results suggest that a subset of lung cancer patients are likely to be more susceptible to DNA cross-link based therapy, or to treatments in which additional repair mechanisms are targeted. These subjects can be identified through FATSI analysis. Clinical trials to evaluate this therapeutic concept are needed. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
7. Phase I trial of non-cytotoxic suramin as a modulator of docetaxel and gemcitabine therapy in previously treated patients with non-small cell lung cancer.
- Author
-
Lam, Elaine T., Au, Jessie L.-S., Otterson, Gregory A., Wientjes, M. Guillaume, Ling Chen, Shen, Tong, Yong Wei, Xiaobai Li, Bekii-Saab, Tanios, Murgo, Anthony J., Jensen, Rhonda R., Grever, Michael, and Villalona-Calero, Miguel A.
- Subjects
ANTINEOPLASTIC agents ,DRUG therapy ,LUNG cancer ,MEDICAL research ,QUANTITATIVE research - Abstract
Purpose: In preclinical models, non-cytotoxic suramin (concentrations <50 μM) potentiates the activity of multiple chemotherapeutic agents. The present study evaluated the safety and tolerability of suramin in combination with docetaxel or gemcitabine in previously chemotherapy-treated patients with advanced non-small cell lung cancer. Methods: Patients received suramin intravenously in combination with either docetaxel on day 1 or gemcitabine on days 1 and 8, of each 21-day treatment cycle. After 3 cycles, patients with partial response (PR) or better continued on the same combination, whereas patients with stable disease (SD) or worse crossed-over to the other combination. Pharmacokinetic analyses were performed before and after each treatment. Results: Eighteen patients received a total of 79 courses (37 suramin plus docetaxel, 42 suramin plus gemcitabine). The dose-limiting toxicity (DLT) was febrile neutropenia, observed in three of six patients treated with suramin and docetaxel 75 mg/m. No DLTs were observed with suramin plus docetaxel 56 mg/m or suramin plus gemcitabine 1,250 mg/m. Common adverse events included neutropenia, thrombocytopenia, anemia, fatigue, nausea, vomiting, skin rash, hyperglycemia, and electrolyte abnormalities. The target plasma suramin concentration range of 10-50 μM was achieved in 90% of treatments. Discernable antitumor activity was noted in 11 patients (2 PR, 9 SD). Conclusions: Non-cytotoxic suramin, in combination with docetaxel 56 mg/m or gemcitabine 1,250 mg/m, was reasonably well-tolerated with a manageable toxicity profile. Target plasma concentrations were correctly predicted by our previously described dosing nomogram. The observed preliminary evidence of antitumor activity encourages evaluation of this strategy in efficacy trials. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
8. Expression of a Mutant p53 Results in an Age-Related Demographic Shift in Spontaneous Lung Tumor Formation in Transgenic Mice.
- Author
-
Wenrui Duan, Li Gao, Xin Wu, Hade, Erinn M., Jian-Xin Gao, Haiming Ding, Barsky, Sanford H., Otterson, Gregory A., and Villalona-Calero, Miguel A.
- Subjects
LUNG cancer ,TRANSGENIC mice ,TUMOR growth ,HISTOPATHOLOGY ,NUCLEIC acids ,METHYLATION ,ALKYLATION ,ANIMAL morphology ,ONCOLOGY ,DIAGNOSIS - Abstract
Background: Mutations in the P53 gene are among the most common genetic abnormalities in human lung cancer. Codon 273 in the sequence-specific DNA binding domain is one of the most frequently mutated sites. Methodology: To investigate the role of mutant p53 in lung tumorigenesis, a lung specific p53(273H) transgenic mouse model was developed. Rates of lung cancer formation in the transgenic animals and their littermates were evaluated by necropsy studies performed in progressive age cohorts ranging from 4 to 24 months. In order to establish the influence of other common genetic abnormalities in lung tumor formation in the animals, K-Ras gene mutation and p16INK4a (p16) promoter methylation were evaluated in a total of 281 transgenic mice and 189 non-transgenic littermates. Principal Findings: At the age extremes of 4-12 and 22-24 months no differences were observed, with very low prevalence of tumors in animals younger than 12 months, and a relatively high prevalence at age 22 months or older. However, the transgenic mice had a significant higher lung tumor rate than their non-transgenic counterparts during the age of 13-21 months, suggesting an age-related shift in lung tumor formation induced by the lung-specific expression of the human mutant p53. Histopathology suggested a more aggressive nature for the transgenic tumors. Older mice (>13 months) had a significantly higher rate of p16 promoter methylation (17% v 82%). In addition, an age related effect was observed for K-Ras codons 12 or 13 mutations, but not for codon 61 mutations. Conclusions/Significance: These results would suggest that the mutant p53(273H) contributes to an acceleration in the development of spontaneous lung tumors in these mice. Combination with other genetic and epigenetic alterations occurring after the age of 13 months is intimately linked to its oncogenic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
9. Expression of Pirh2, a newly identified ubiquitin protein ligase, in lung cancer.
- Author
-
Duan, Wenrui, Li Gao, Druhan, Lawrence J., Zhu, Wei-Guo, Morrison, Carl, Otterson, Gregory A., Villalona-Calero, Miguel A., and Gao, Li
- Subjects
LIGASES ,UBIQUITIN ,PROTEINS ,LUNG cancer ,PROTEOMICS ,ONCOLOGY ,ENZYME analysis ,PROTEIN metabolism ,ENZYME metabolism ,ANIMAL experimentation ,BIOCHEMISTRY ,COMPARATIVE studies ,GENES ,IMMUNOBLOTTING ,IMMUNOHISTOCHEMISTRY ,LUNGS ,LUNG tumors ,PHENOMENOLOGY ,RESEARCH methodology ,MEDICAL cooperation ,MICE ,POLYMERASE chain reaction ,RESEARCH ,EVALUATION research ,REVERSE transcriptase polymerase chain reaction - Abstract
Maintenance of p53 function is important for normal cell growth and development, and loss of p53 function contributes directly to malignant tumor development. The recently discovered Pirh2 protein is an ubiquitin-protein ligase that negatively regulates p53 through activity by targeting it for degradation. To determine how Pirh2 may mediate lung tumorigenesis, we evaluated Pirh2 expression in human and mouse lung tumor samples and paired normal lung tissues using immunoblot analysis and immunohistochemistry. Pirh2 protein expression was higher in 27 (84%) of 32 human lung neoplasms than in matched normal lung tissue and in 14 of 15 mouse lung tumors evaluated. In addition, p53 protein was more ubiquitinated in the mouse lung tumors than in normal tissue, and overall p53 expression was lower than that in normal tissue. These results are consistent with the hypothesis that increased Pirh2 expression affects lung tumorigenesis by reducing p53 activity. To our knowledge, this is the first description of altered Pirh2 expression in human and mouse tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
10. Increased expression of unmethylated CDKN2D by 5-aza-2′-deoxycytidine in human lung cancer cells.
- Author
-
Zhu, Wei-Guo, Dai, Zunyan, Ding, Haiming, Srinivasan, Kanur, Hall, Julia, Duan, Wenrui, Villalona-Calero, Miguel A, Plass, Christoph, and Otterson, Gregory A
- Subjects
LUNG cancer ,CANCER cells ,METHYLATION - Abstract
DNA hypermethylation of CpG islands in the promoter region of genes is associated with transcriptional silencing. Treatment with hypo-methylating agents can lead to expression of these silenced genes. However, whether inhibition of DNA methylation influences the expression of unmethylated genes has not been extensively studied. We analysed the methylation status of CDKN2A and CDKN2D in human lung cancer cell lines and demonstrated that the CDKN2A CpG island is methylated, whereas CDKN2D is unmethylated. Treatment of cells with 5-aza-2′-deoxycytidine (5-Aza-CdR), an inhibitor of DNA methyltransferase 1, induced a dose and duration dependent increased expression of both p16
INK4a and p19INK4d , the products of CDKN2A and CDKN2D, respectively. These data indicate that global DNA demethylation not only influences the expression of methylated genes but also of unmethylated genes. Histone acetylation is linked to methylation induced transcriptional silencing. Depsipeptide, an inhibitor of histone deacetylase, acts synergistically with 5-Aza-CdR in inducing expression of p16INK4a and p19INK4d . However, when cells were treated with higher concentrations of 5-Aza-CdR and depsipeptide, p16INK4a expression was decreased together with significant suppression of cell growth. Interestingly, p19INK4d expression was enhanced even more by the higher concentrations of 5-Aza-CdR and depsipeptide. Our data suggest that p19INK4d plays a distinct role from other INK4 family members in response to the cytotoxicity induced by inhibition of DNA methylation and histone deacetylation. Oncogene (2001) 20, 7787–7796. [ABSTRACT FROM AUTHOR]- Published
- 2001
- Full Text
- View/download PDF
11. Differential response between the p53 ubiquitin–protein ligases Pirh2 and MdM2 following DNA damage in human cancer cells
- Author
-
Duan, Wenrui, Gao, Li, Wu, Xin, Zhang, Yang, Otterson, Gregory A., and Villalona-Calero, Miguel A.
- Subjects
- *
PROTEINS , *CANCER , *TUMORS , *DNA damage - Abstract
Abstract: Pirh2, a recently identified ubiquitin–protein ligase, has been reported to promote p53 degradation. Pirh2 physically interacts with p53 and promotes ubiquitination of p53 independently of MDM2. Like MDM2, Pirh2 is thought to participate in an autoregulatory feedback loop that controls p53 function. We have previously reported that Pirh2 was overexpressed in human and murine lung cancers as compared to uninvolved lung tissue. Pirh2 increase could potentially cause degradation of wildtype p53 and reduce its tumor suppression function in the lung tumor cells. Since Pirh2 has been reported to be transactivated by p53, however, the mechanisms by which a high level of Pirh2 expression is maintained in tumor cells despite low level of wildtype p53 protein are unclear. In order to evaluate p53 involvement in the transactivation of Pirh2, we evaluated Pirh2, MDM2, p53 and p21 expression with Western blot analysis and real time PCR after γ irradiation or cisplatin DNA damage treatment using human cancer cell lines containing wildtype (A549, MCF-7), mutant (H719) and null (H1299) p53. Surprisingly, Pirh2 expression was not affected by the presence of wildtype p53 in the cancer cells. In contrast, MDM2 was upregulated by wildtype p53 in A549 and MCF-7 cells and was absent from the H1299 and the H719 cells. We conclude that Pirh2 operates in a distinct manner from MDM2 in response to DNA damage in cancer cells. Pirh2 elevation in p53 null cells indicates the existence of additional molecular mechanisms for Pirh2 upregulation and suggests that p53 is not the sole target of Pirh2 ubiquitin ligase activity. [Copyright &y& Elsevier]
- 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.