Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 109(47): 19403-7, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23129637

ABSTRACT

No conventional therapy exists for salivary hypofunction in surviving head and neck cancer patients with Radiation Therapy Oncology Group late grade 2-3 toxicity. We conducted a phase I clinical trial to test the safety and biologic efficacy of serotype 5, adenoviral-mediated aquaporin-1 cDNA transfer to a single previously irradiated parotid gland in 11 subjects using an open label, single-dose, dose-escalation design (AdhAQP1 vector; four dose tiers from 4.8 × 10(7) to 5.8 × 10(9) vector particles per gland). Treated subjects were followed at scheduled intervals. Multiple safety parameters were measured and biologic efficacy was evaluated with measurements of parotid salivary flow rate. Symptoms were assessed with a visual analog scale. All subjects tolerated vector delivery and study procedures well over the 42-d study period reported. No deaths, serious adverse events, or dose-limiting toxicities occurred. Generally, few adverse events occurred, and all were considered mild or moderate. No consistent changes were found in any clinical chemistry and hematology parameters measured. Objective responses were seen in six subjects, all at doses <5.8 × 10(9) vector particles per gland. Five of these six subjects also experienced subjective improvement in xerostomia. AdhAQP1 vector delivery to a single parotid gland was safe and transfer of the hAQP1 cDNA increased parotid flow and relieved symptoms in a subset of subjects.


Subject(s)
Adenoviridae/genetics , Aquaporin 1/genetics , Aquaporin 1/therapeutic use , DNA, Complementary/genetics , Genetic Therapy , Radiation Injuries/therapy , Salivary Gland Diseases/therapy , Aged , Citrates , Gallium , Genetic Therapy/adverse effects , Humans , Male , Middle Aged , Radiation Injuries/diagnostic imaging , Radiation Injuries/genetics , Radionuclide Imaging , Salivary Gland Diseases/diagnostic imaging , Salivary Gland Diseases/etiology , Salivary Gland Diseases/physiopathology
2.
Cancer Res ; 71(21): 6867-77, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21933882

ABSTRACT

Inflammation-induced activation of proto-oncogenic NF-κB/REL and dysfunction of tumor suppressor TP53/p63/p73 family transcription factors are key events in cancer progression. How inflammatory signaling coordinates dysregulation of these two transcription factor families during oncogenesis remains incompletely understood. Here, we observed that oncoprotein c-REL and tumor suppressor TAp73 are coexpressed and complex with ΔNp63α in the nucleus of a subset of head and neck squamous cell carcinoma (HNSCC) cell lines with mutant (mt)TP53. TNF-α, a proinflammatory cytokine, promoted c-REL nuclear translocation, c-REL/ΔNp63α interaction, and dissociation of TAp73 from ΔNp63α and the nucleus to the cytoplasm, whereas c-REL siRNA knockdown attenuated this effect. Overexpression of c-REL or a c-REL κB-site DNA-binding mutant enhanced protein interaction with ΔNp63α and TAp73 dissociation, implicating c-REL/ΔNp63α-specific interactions in these effects. We discovered that TNF-α or genetic alteration of c-REL expression inversely modulates ΔNp63α/TAp73 interactions on distinct p63 DNA-binding sites, including those for key growth arrest and apoptotic genes p21WAF1, NOXA, and PUMA. Functionally, c-REL repressed these genes and the antiproliferative effects of TNF-α or TAp73. Conversely, c-REL siRNA depletion enhanced TAp73 promoter interaction and expression of genes mediating growth arrest and apoptosis. Similar to TNF-α-treated HNSCC lines, human HNSCC tumors and hyperplastic squamous epithelia of transgenic mice overexpressing ΔNp63α that exhibit inflammation also show increased nuclear c-REL/ΔNp63α and cytoplasmic TAp73 localization. These findings unveil a novel and reversible dynamic mechanism whereby proinflammatory cytokine TNF-α-induced c-REL/ΔNp63α interactions inactivate tumor suppressor TAp73 function, promoting TNF-α resistance and cell survival in cancers with mtTP53.


Subject(s)
Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Head and Neck Neoplasms/pathology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Binding Sites , Carcinoma, Squamous Cell/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Neoplasm/metabolism , Drug Resistance/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, p53 , Genes, rel , Head and Neck Neoplasms/genetics , Humans , Multiprotein Complexes , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-rel , RNA, Small Interfering/pharmacology
3.
Int J Radiat Oncol Biol Phys ; 77(2): 447-54, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-19879702

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) overexpression in head-and-neck squamous cell carcinoma (HNSCC) stimulates tumor cell proliferation, inhibits apoptosis, and increases chemotherapy and radiation resistance. We examined the toxicity, safety and the effects on EGFR signaling in tumor biopsy samples from patients with locally advanced HNSCC treated with the EGFR signaling inhibitor gefitinib (GEF) combined with weekly intravenous paclitaxel (PAC) and radiation therapy (RT). METHODS AND MATERIALS: This was a pilot Phase I dose-escalation study. Eligibility included Stage III to IVB HNSCC, age >or=18 years, no prior RT or chemotherapy, adequate organ function, and informed consent. Endpoints included determination of maximum tolerated dose (MTD) and analysis of treatment effect on EGFR signaling, tumor cell proliferation, and apoptosis in biopsy samples. RESULTS: Ten patients were treated. The MTD of this combination was GEF 250 mg/d with PAC 36 mg/m(2) intravenously weekly x 6 with concurrent RT. Grade 3/4 toxicities included prolonged (>8 weeks) stomatitis (7 patients), infection (2 patients), and interstitial pneumonitis (1 patient). There were five complete responses (CR) and two partial responses (PR). Of 7 patients undergoing serial biopsies, only 1 patient demonstrated a reduction in phosphorylated EGFR, decreased downstream signaling, and reduced cellular proliferation after initiating GEF. CONCLUSIONS: Inhibition of EGFR by GEF was observed in only one of seven tumors studied. The addition of GEF to PAC and RT did not appear to improve the response of locally advanced HNSCC compared with our prior experience with PAC and RT alone. This treatment appeared to delay recovery from stomatitis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Drug Administration Schedule , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Female , Gefitinib , Head and Neck Neoplasms/pathology , Humans , Infections/etiology , Lung Diseases, Interstitial/etiology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Pilot Projects , Quinazolines/administration & dosage , Quinazolines/adverse effects , Radiotherapy Dosage , Remission Induction , Stomatitis/etiology
4.
Mol Cancer Ther ; 7(7): 1949-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645005

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) exhibit constitutive activation of transcription factors nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1), which are modulated by the proteasome and promote resistance to cell death. HNSCC show variable sensitivity to the proteasome inhibitor bortezomib in vitro as well as in murine xenografts and patient tumors in vivo, and the mechanisms are not well understood. To address this question, the sensitivities of nine HNSCC cell lines to bortezomib were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, and the potential relationship between the sensitivity and bortezomib effects on biological processes was examined in HNSCC lines of differential bortezomib sensitivity. The most sensitive cell line (UM-SCC-11B) underwent cell death at 10(-9) mol/L in vitro and tumor regression at a maximally tolerated dose of bortezomib in a murine xenograft model. The differential sensitivity between UM-SCC-11A and UM-SCC-11B cells corresponded to differences in the extent of suppression of proteasome activity, ubiquitinated protein degradation, and NF-kappaB and AP-1 activation. Lower concentrations of bortezomib transiently increased NF-kappaB and sustained AP-1 activation in UM-SCC-11A cells. AP-1 reporter activity and cell density of UM-SCC-11A were suppressed when bortezomib was combined with c-Jun NH(2)-terminal kinase and p38 kinase pathways inhibitors. Thus, the differential sensitivities to bortezomib corresponded to dissimilar effects on the proteasome, NF-kappaB and AP-1 activities. Inhibition of c-Jun NH(2)-terminal kinase and p38 pathways blocked AP-1 activity and enhanced the antitumor effects. These findings revealed molecular mechanisms of bortezomib sensitivity and resistance, which are under development as biomarkers for clinical trials in patients with HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Head and Neck Neoplasms/pathology , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Transcription Factor AP-1/metabolism , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Bortezomib , Cell Cycle/drug effects , Cell Line, Tumor , Drug Synergism , Genes, Reporter , Humans , Keratinocytes/drug effects , Mice , Mice, Inbred BALB C , Proteasome Inhibitors , Signal Transduction/drug effects , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 14(13): 4175-85, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18593997

ABSTRACT

PURPOSE: Nuclear factor-kappaB (NF-kappaB)/REL transcription factors promote cancer cell survival and progression. The canonical (NF-kappaB1/RELA or cREL) and alternate (NF-kappaB2/RELB) pathways require the proteasome for cytoplasmic-nuclear translocation, prompting the investigation of bortezomib for cancer therapy. However, limited clinical activity of bortezomib has been observed in many epithelial malignancies, suggesting this could result from incomplete inhibition of NF-kappaB/RELs or other prosurvival signal pathways. EXPERIMENTAL DESIGN: To examine these possibilities, matched biopsies from 24 h posttreatment were obtained from accessible tumors of patients who received low-dose bortezomib (0.6 mg/m(2)) before reirradiation in a phase I trial for recurrent head and neck squamous cell carcinoma (HNSCC). Effects of bortezomib on apoptosis and proliferation by TUNEL and Ki67 staining were compared with nuclear staining for all five NF-kappaB subunits, phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated signal transducers and activators of transcription 3 (STAT3) in tumor biopsies, and by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTP) and DNA binding assay for the five NF-kappaB subunits in HNSCC cell lines. RESULTS: HNSCC showed increased nuclear staining for all five NF-kappaB subunits, phosphorylated ERK1/2, and phosphorylated STAT3. Bortezomib treatment significantly enhanced apoptosis with inhibition of nuclear RELA in three of four tumors, but other NF-kappaB subunits, ERK1/2, and STAT3 were variably or not affected, and tumor progression was observed within 3 months. In HNSCC cell lines, 10(-8) mol/L bortezomib inhibited cell density while inhibiting tumor necrosis factor-alpha-induced and partially inhibiting basal activation of NF-kappaB1/RELA, but not NF-kappaB2/RELB. CONCLUSIONS: Although low-dose bortezomib inhibits activation of subunits of the canonical pathway, it does not block nuclear activation of the noncanonical NF-kappaB or other prosurvival signal pathways, which may contribute to the heterogeneous responses observed in HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Boronic Acids/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , NF-kappa B/metabolism , Pyrazines/pharmacology , Bortezomib , Cell Nucleus/metabolism , DNA/chemistry , Humans , Models, Biological , Phosphorylation , Recurrence , Signal Transduction , Subcellular Fractions/metabolism
6.
Clin Cancer Res ; 13(22 Pt 1): 6568-78, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006756

ABSTRACT

PURPOSE: To determine the nature and potential pharmacologic reversibility of deficient TP53 expression and function in head and neck squamous cell carcinomas (HNSCC) with wild-type TP53, previously associated with decreased sensitivity to cisplatin therapy. EXPERIMENTAL DESIGN: TP53 genotype, mRNA and protein expression, TP53-induced p21 expression, and TP53 DNA-binding and reporter gene function were determined in a panel of nine previously characterized HNSCC cell lines from the University of Michigan squamous cell carcinoma (UM-SCC) series. The genotoxic drug doxorubicin and the anti-inflammatory and antimalarial drug quinacrine, previously identified as inducers of TP53, were used to examine the nature and potential reversibility of deficient TP53 expression and function. The specific role of inducible TP53 on function and cellular proliferation was confirmed using selective TP53 inhibitor pifithrin-alpha or short hairpin RNA knockdown. The capability of quinacrine to sensitize HNSCC to the cytotoxic effects of cisplatin was assessed. RESULTS: UM-SCC cell lines with wild-type TP53 genotype underexpressed TP53 mRNA and protein when compared with normal human keratinocytes or UM-SCC with mutant TP53. Although doxorubicin failed to induce TP53 expression or functional activity, quinacrine induced TP53 mRNA and protein expression, increased TP53 reporter activity and p21 protein expression, and induced growth inhibition in these wild-type TP53 cell lines. Quinacrine-induced TP53 reporter activity and growth suppression were attenuated by pifithrin-alpha and TP53 short hairpin RNA knockdown. Furthermore, quinacrine sensitized UM-SCC to cisplatin in vitro. CONCLUSIONS: Deficient TP53 mRNA and protein expression underlies decreased function in a subset of HNSCC with wild-type TP53 and can be restored together with cisplatin sensitization by quinacrine.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/drug therapy , Quinacrine/therapeutic use , Tumor Suppressor Protein p53/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Head and Neck Neoplasms/pathology , Humans , Quinacrine/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics
8.
Mol Cancer Ther ; 6(1): 37-50, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237265

ABSTRACT

Histone deacetylase inhibitors (HDI) can inhibit proliferation and enhance apoptosis in a wide range of malignancies. However, HDIs show relatively modest activity in head and neck squamous cell carcinomas (HNSCC), in which we have shown the activation of nuclear factor-kappaB (NF-kappaB; NF-kappaB1/RelA or p50/p65), a transcription factor that promotes expression of proliferative and antiapoptotic genes. In this study, we examined if HDIs enhance activation of NF-kappaB and target genes and if genetic or pharmacologic inhibition of NF-kappaB can sensitize HNSCC to HDIs. Limited activity of classic HDIs trichostatin A and sodium butyrate was associated with enhanced activation of NF-kappaB reporter activity in a panel of six HNSCC cell lines. HDIs enhanced NF-kappaB p50/p65 DNA binding and acetylation of the RelA p65 subunit. Transfection of small interfering RNAs targeting p65 strongly inhibited NF-kappaB expression and activation, induced cell cycle arrest and cell death, and further sensitized HNSCC cells when combined with HDIs. The p65 small interfering RNA inhibited HDI-enhanced expression of several NF-kappaB-inducible genes implicated in oncogenesis of HNSCC, such as p21, cyclin D1, and BCL-XL. Bortezomib, an inhibitor of proteasome-dependent NF-kappaB activation, also increased sensitization to trichostatin A, sodium butyrate, and a novel HDI, PXD101, in vitro, and to the antitumor effects of PXD101 in bortezomib-resistant UMSCC-11A xenografts. However, gastrointestinal toxicity, weight loss, and mortality of the combination were dose limiting and required parenteral fluid administration. We conclude that HDI-enhanced NF-kappaB activation is one of the major mechanisms of resistance of HNSCC to HDIs. The combination of HDI and proteasome inhibitor produced increased antitumor activity. Low starting dosages for clinical studies combining HDIs with proteasome inhibitors and IV fluid support may be warranted.


Subject(s)
Boronic Acids/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Hydroxamic Acids/therapeutic use , Proteasome Inhibitors , Pyrazines/therapeutic use , RNA, Small Interfering/metabolism , Transcription Factor RelA/metabolism , Acetylation/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Boronic Acids/administration & dosage , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Squamous Cell/pathology , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, SCID , NF-kappa B p50 Subunit/metabolism , Protein Binding/drug effects , Pyrazines/administration & dosage , Pyrazines/pharmacology , Sulfonamides , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
9.
Curr Opin Otolaryngol Head Neck Surg ; 12(4): 323-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15252255

ABSTRACT

PURPOSE OF REVIEW: Since the theory of selective photothermolysis was described, the field of facial plastic surgery has advanced the science behind laser: tissue interactions. Particular advances in skin cooling, hair removal, intense pulsed light, and uses for aesthetic and nonaesthetic skin problems are described in this review. RECENT FINDINGS: Continued basic science research in lasers has led to improvements in current technology available to the facial plastic surgeon. Advances in skin cooling have allowed for wider use in all Fitzpatrick skin types without concomitant adverse reaction. SUMMARY: Our understanding of laser science continues to expand our knowledge in basic laser: tissue interaction. An improvement in various laser mediums and adjunctive devices provides the facial plastic surgeon with instruments to treat a wider patient population. The benefit is a successful clinical and aesthetic result with improved safety.


Subject(s)
Face/surgery , Laser Therapy/methods , Surgery, Plastic/methods , Cold Temperature , Dermatologic Surgical Procedures , Hair Removal/methods , Humans , Laser Therapy/standards , Skin Pigmentation , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...