Searched for: person:pavlia01
Phase I study of bryostatin 1, a protein kinase C modulator, preceding cisplatin in patients with refractory non-hematologic tumors
Pavlick, Anna C; Wu, Jennifer; Roberts, John; Rosenthal, Mark A; Hamilton, Anne; Wadler, Scott; Farrell, Kathleen; Carr, Michelle; Fry, David; Murgo, Anthony J; Oratz, Ruth; Hochster, Howard; Liebes, Leonard; Muggia, Franco
PURPOSE: Preclinical data suggested that bryostatin-1 (bryo) could potentiate the cytotoxicity of cisplatin when given prior to this drug. We designed a phase I study to achieve tolerable doses and schedules of bryo and cisplatin in combination and in this sequence. METHODS: Patients with non-hematologic malignancies received bryo followed by cisplatin in several schedules. Bryo was given as an 1 and a 24 h continuous infusion, while cisplatin was always given over 1 h at 50 and 75 mg/m(2); the combined regimen was repeated on an every 3-week and later on an every 2-week schedule. Bryo doses were escalated until recommended phase II doses were defined for each schedule. Patients were evaluated with computerized tomography every 2 cycles. RESULTS: Fifty-three patients were entered. In an every 2-week schedule, the 1-h infusion of bryo became limited by myalgia that was clearly cumulative. With cisplatin 50 mg/m(2) its recommended phase II dose was 30 mug/m(2). In the 3-week schedule, dose-limiting toxicities were mostly related to cisplatin effects while myalgias were tolerable. Pharmacokinetics unfortunately proved to be unreliable due to bryo's erratic extraction. Consistent inhibition of PKC isoform eta (eta) in peripheral blood mononuclear cells was observed following bryo. CONCLUSIONS: Bryo can be safely administered with cisplatin with minimal toxicity; however, only four patients achieved an objective response. Modulation of cisplatin cytotoxicity by bryo awaits further insight into the molecular pathways involved
PMCID:3901370
PMID: 19221754
ISSN: 1432-0843
CID: 97002
Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features
Creighton, Chad J; Li, Xiaoxian; Landis, Melissa; Dixon, J Michael; Neumeister, Veronique M; Sjolund, Ashley; Rimm, David L; Wong, Helen; Rodriguez, Angel; Herschkowitz, Jason I; Fan, Cheng; Zhang, Xiaomei; He, Xiaping; Pavlick, Anne; Gutierrez, M Carolina; Renshaw, Lorna; Larionov, Alexey A; Faratian, Dana; Hilsenbeck, Susan G; Perou, Charles M; Lewis, Michael T; Rosen, Jeffrey M; Chang, Jenny C
Some breast cancers have been shown to contain a small fraction of cells characterized by CD44(+)/CD24(-/low) cell-surface antigen profile that have high tumor-initiating potential. In addition, breast cancer cells propagated in vitro as mammospheres (MSs) have also been shown to be enriched for cells capable of self-renewal. In this study, we have defined a gene expression signature common to both CD44(+)/CD24(-/low) and MS-forming cells. To examine its clinical significance, we determined whether tumor cells surviving after conventional treatments were enriched for cells bearing this CD44(+)/CD24(-/low)-MS signature. The CD44(+)/CD24(-/low)-MS signature was found mainly in human breast tumors of the recently identified "claudin-low" molecular subtype, which is characterized by expression of many epithelial-mesenchymal-transition (EMT)-associated genes. Both CD44(+)/CD24(-/low)-MS and claudin-low signatures were more pronounced in tumor tissue remaining after either endocrine therapy (letrozole) or chemotherapy (docetaxel), consistent with the selective survival of tumor-initiating cells posttreatment. We confirmed an increased expression of mesenchymal markers, including vimentin (VIM) in cytokeratin-positive epithelial cells metalloproteinase 2 (MMP2), in two separate sets of postletrozole vs. pretreatment specimens. Taken together, these data provide supporting evidence that the residual breast tumor cell populations surviving after conventional treatment may be enriched for subpopulations of cells with both tumor-initiating and mesenchymal features. Targeting proteins involved in EMT may provide a therapeutic strategy for eliminating surviving cells to prevent recurrence and improve long-term survival in breast cancer patients.
PMCID:2720409
PMID: 19666588
ISSN: 0027-8424
CID: 421022
Phase II assessment of talabostat and cisplatin in second-line stage IV melanoma
Eager, Robert M; Cunningham, C Casey; Senzer, Neil N; Stephenson, Joe Jr; Anthony, Stephen P; O'Day, Steven J; Frenette, Gary; Pavlick, Anna C; Jones, Barry; Uprichard, Margaret; Nemunaitis, John
BACKGROUND: Metastatic melanoma is an incurable disease with an average survival of less than one year. Talabostat is a novel dipeptidyl peptidase inhibitor with immunostimulatory properties. METHODS: This phase II, open label, single arm study was conducted to evaluate the safety and efficacy of 75-100 mg/m2 cisplatin combined with 300-400 mcg talabostat bid for 6, 21-day cycles. The primary endpoint was overall response. The rate of complete responses, duration of overall objective response, progression-free survival (PFS), and overall survival were the secondary endpoints. RESULTS: Six objective partial responses were recorded in the 74 patients (8.1%) in the intention-to-treat population. Five of these responses involved the 40 evaluable patients (12.5%). Thirty-one percent of patients reported SAEs to the combination of talabostat and cisplatin. CONCLUSION: Acceptable tolerability was observed in the intention-to-treat population and antitumor activity was observed in 12.5% of evaluable patients, which is not greater than historical expectation with cisplatin alone
PMCID:2731782
PMID: 19643020
ISSN: 1471-2407
CID: 141639
Novel therapeutics for melanoma
Seetharamu, Nagashree; Ott, Patrick A; Pavlick, Anna C
Advanced melanoma has the highest per-death loss of years of potential life expectancy except for adult leukemia. Standard therapy with agents such as dacarbazine, temozolomide and IL-2 is associated with notoriously low response rates. The identification of new active agents is, therefore, critical in this disease. In recent years, better understanding of melanoma biology, as well as cancer and immune biology in general has led to the development of a number of new potential therapeutic agents for advanced melanoma. While many of these compounds are being tested in clinical trials, there are more agents in various stages of preclinical development. These novel therapeutics offer hope for this aggressive and so far largely treatment-resistant disease. In this review we will discuss some of the most promising novel therapeutic agents for advanced melanoma
PMID: 19496721
ISSN: 1744-8328
CID: 99325
A phase I/II study of CR011-vcMMAE, an antibody-drug conjugate (ADC) targeting glycoprotein NMB (GPNMB) in patients (pts) with advanced melanoma [Meeting Abstract]
Hwu, P.; Sznol, M.; Pavlick, A.; Kluger, H.; Kim, K. B.; Boasberg, P.; Sanders, D.; Simantov, R.; Crowley, E.; Hamid, O.
ISI:000276606606048
ISSN: 0732-183x
CID: 3158932
Effect of mebendazole on melanoma xenograft growth through targeting of bcl-2 [Meeting Abstract]
Doudican, N. A.; Pennell, R.; Tu, T.; Liebes, L.; Pavlick, A.; Berman, R.; Shapiro, R.; Goldberg, J. D.; Osman, I.; Orlow, S.
ISI:000276606606090
ISSN: 0732-183x
CID: 3159012
Use of gene expression profile and mitotic index of metastatic melanoma lesions as an adjunct to TNM staging in predicting patient survival [Meeting Abstract]
Bogunovic, D.; O'Neill, D.; Belitskaya-Levy, I.; Vacic, V.; Adams, S.; Darvishian, F.; Pavlick, A.; Zavadil, J.; Osman, I.; Bhardwaj, N.
ISI:000276606606030
ISSN: 0732-183x
CID: 3159052
Detection of BRAF kinase mutations in melanoma, ovarian, and prostate carcinomas: Evidence for tumor heterogeneity in clinical samples [Meeting Abstract]
Litterman, A J; Yancovitz, M; Shapiro, R; Berman, R; Pavlick, A; Daarvishian, F; Blank, S; Lee, P; Osman, I; Polsky, D
Background: Several studies have provided evidence that solid tumors are polyclonal malignancies, an observation which may contribute to difficulties in achieving durable treatment responses. In some patients, molecularly targeted therapies may be compromised due to heterogeneity among tumor subclones. In this study we compared conventional DNA sequencing with a fluorescent-based mutant-specific PCR (MS-PCR) assay to detect the BRAF hotspot mutation V600E in a large panel of patient tumors, including paired primary and metastatic tumors from individual patients. Methods: BRAF MS-PCR and conventional sequencing were performed on DNA from 304 tumors (112 melanoma, 110 ovarian, 82 prostate) to determine the presence of the BRAFV600E hot-spot mutation. Among the melanomas were 18 matched primary and metastatic specimens, and 40 metastatic specimens from 19 patients, each of whom had 2 or more metastases. Results: DNA sequencing detected mutations in 5/110 (4.5%) ovarian tumors, 1/82 (1.2%) prostate tumors, and 36/112 (32%) melanomas. In contrast, the MS-PCR assay detected mutations in 12/110 (11%) ovarian tumors, 15/82 (18%) prostate tumors and 85/112 (76%) melanomas. The presence of contaminating normal tissue was scored for each melanoma sample, but excess normal tissue did not influence the results using either methodology. In all cases mutations detected by sequencing were also detected by MSPCR. Among 18 patients with matched primary and metastatic melanoma, 8/18 (44%) had discordant results including 2 patients with mutant primary tumors and wild-type metastases; among the 19 patients with multiple metastases 5/19 (26%) had discordant (both wild-type and mutant) tumors. Conclusions: Using a highly sensitive BRAF mutation detection method, we observed substantial evidence for heterogeneity within clinical tumor specimens. This was especially true in melanoma samples, where multiple specimens from individual patients differed with respect to the presence of the mutant BRAF allele. These results suggest that failures of molecularly targeted therapies, such as those directed against mutant BRAF, may be due in part to a lack of clonality among the tumors under treatment
EMBASE:70243349
ISSN: 0732-183x
CID: 3159892
Skp2, a prognostic marker and potential therapeutic target in metastatic melanoma [Meeting Abstract]
Wang, G; Hanniford, D; Rose, A; Gaziel, A; Pavlick, A; Zhou, X; Wang, J; Pagano, M; Hernando, E; Osman, I
Background: Skp2, a known oncogene, is overexpressed in several types of tumors and is associated with worse recurrence rate and overall survival in primary melanoma patients. Moreover, the anti-proliferative effects of Skp2 siRNA on various tumor cell lines have prompted the preclinical testing of Skp2 small molecule inhibitors. In this study, we assessed the clinical relevance and molecular mechanism(s) underlying Skp2 overexpression in metastatic melanoma patients. Methods: Skp2 protein levels were measured in 122 metastatic melanoma specimens using immunohistochemistry (IHC), and the association between Skp2 overexpression and post-recurrence survival was examined. Moreover, 22 cell lines (2 normal primary melanocytes, 2 primary immortal melanocytes, 4 primary melanoma cell lines, and 18 metastatic melanoma cell lines) were evaluated for Skp2 genomic amplification using Single Nucleotide Polymorphism (SNP) arrays (Affymetrix 6.0) and Skp2 gene expression using mRNA arrays (Affymetrix U133A 2.0) and quantitative RT-PCR. We also screened 18 cell lines for Skp2 mutation by sequencing. Results: Skp2 overexpression, defined as >25% tumor cells, was associated with shorter 3-yr post-recurrence survival (37%) compared to Skp2 expression <= 25% (55%) (HR=1.89, 95%, CI= 1.04, 3.42, p=0.04). Skp2 overexpression was significantly associated with the site of melanoma metastasis: visceral (n= 12; 89%), lymph node (n=49; 36%), brain (n=15; 14%), and soft-tissue (n=36; 6%) (p<0.001). SNP array revealed genomic amplification at the Skp2 locus in 6 (33%) metastatic cell lines and one primary melanoma cell line. Skp2 genomic amplification was associated with increased transcript expression. No Skp2 mutations were identified. Conclusions: Skp2 protein overexpression is associated with worse prognosis in metastasis in melanoma. Our results also support that gene amplification, rather than a Skp2 gene mutation, may be the major mechanism responsible for Skp2 aberrant expression in metastatic melanoma
EMBASE:70243352
ISSN: 0732-183x
CID: 3159952
Prospective analysis of predictors of survival in melanoma patients with brain metastases [Meeting Abstract]
Zakrzewski, J. A.; Geraghty, L.; Hamilton, H.; Christos, P.; Krich, D.; Mazumdar, M.; Polsky, D.; Darvishian, F.; Pavlick, A.; Osman, I.
ISI:000276606606089
ISSN: 0732-183x
CID: 3159022