Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Correct Health Care ; 24(1): 71-83, 2018 01.
Article in English | MEDLINE | ID: mdl-29303039

ABSTRACT

Incarceration history can affect sexual health behaviors. A randomized controlled trial of a prevention intervention tailored for post-incarcerated men was administered in a reentry setting. Men ≤45 days post release were recruited into a five-session intervention study. Participants ( N = 255) were assessed and tested for three sexually transmitted diseases (STDs) and HIV at baseline and 3 months post-intervention and followed up for 3 more months. The intervention group's STD risks knowledge ( p < .001), partner communication about condoms ( p < .001), and condom application skills ( p < .001) improved. Although fewer men tested positive for an STD at 3 months post-intervention (10% vs. 8%) and no new HIV cases were found, the finding was not significant. A tailored risk reduction intervention for men with incarceration histories can affect sexual risk behaviors.


Subject(s)
Prisons , Sexually Transmitted Diseases/prevention & control , Adolescent , Adult , Condoms/statistics & numerical data , Health Knowledge, Attitudes, Practice , Humans , Interpersonal Relations , Male , Middle Aged , Risk-Taking , Young Adult
2.
Cancer Res ; 68(15): 6232-40, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676847

ABSTRACT

Clinical studies have shown that decreased tamoxifen effectiveness correlates with elevated levels of vascular endothelial growth factor (VEGF)-A(165) in biopsy samples of breast cancers. To investigate the mechanisms underlying tamoxifen resistance and metastasis, we engineered the estrogen receptor (ER)-positive MCF-7 human breast cancer cell line to express VEGF to clinically relevant levels in a doxycycline-regulated manner. Induction of VEGF expression in orthotopically implanted xenografts that were initially tamoxifen responsive and noninvasive resulted in tamoxifen-resistant tumor growth and metastasis to the lungs. Lung metastases were also observed in a VEGF-dependent manner following tail vein injection of tumor cells. At both primary and metastatic sites, VEGF-overexpressing tumors exhibited extensive fibroblastic stromal content, a clinical feature called desmoplasia. VEGF-induced metastatic colonies were surrounded by densely packed stromal cells before detectable angiogenesis, suggesting that VEGF is involved in the initiation of desmoplasia. Because expression of VEGF receptors R1 and R2 was undetectable in these tumor cells, the observed VEGF effects on reduction of tamoxifen efficacy and metastatic colonization are most likely mediated by paracrine signaling that enhances tumor/stromal cell interactions and increases the level of desmoplasia. This study reveals new roles for VEGF in breast cancer progression and suggests that combination of antiestrogens and VEGF inhibitors may prolong tamoxifen sensitivity and prevent metastasis in patients with ER-positive tumors.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Neoplasm Metastasis , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor A/physiology , Humans , Signal Transduction/drug effects
3.
Gynecol Oncol ; 100(3): 521-32, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16225913

ABSTRACT

OBJECTIVE: We have previously shown that adenoviral-mediated melanoma differentiation-associated gene-7 (Ad.mda-7) therapy induces apoptosis in ovarian cancer cells. However, the apoptosis induction was low and directly correlated with infectivity of Ad.mda-7. The objective of this study was to derive ovarian cancer targeted infectivity-enhanced adenoviral vectors encoding mda-7 and evaluate their enhancement in therapeutic efficacy for ovarian carcinoma. METHODS: Infectivity-enhanced adenoviral vectors encoding mda-7 Ad.RGD.mda-7 and Ad.RGD.pK7.mda-7 were derived by incorporation of RGD and or RGD and Pk7 motifs in the fiber knobs by genetic modification. Viruses were validated by PCR for presence of mda-7 and by Western blot for expression of MDA-7 protein. To test the enhancement of therapeutic efficacy of these viruses, a panel of human ovarian carcinoma cells, OV-4, HEY, SKOV3, SKOV3.ip1, were infected by either Ad.mda-7 or Ad.RGD.mda-7 and Ad.RGD.pK7.mda-7 or their respective control viruses and the cell killing was evaluated by crystal violet staining in vitro. Further, therapeutic efficacy was evaluated in vivo using human ovarian cancer xenograft mouse models. RESULTS: Both Ad.RGD.pK7.mda-7 and Ad.RGD.mda-7 showed significant increase in cell killing in vitro compared to unmodified Ad.mda-7 with Ad.RGD.pK7.mda-7 showing highest cell killing. Further, Ad.RGD.pK7.mda-7 showed a significant increase in survival of mice bearing human ovarian cancer xenografts compared to Ad.mda-7 and other control groups. CONCLUSION: Infectivity-enhanced Ad.RGD.mda-7 and Ad.RGD.pK7.mda-7 viruses significantly enhanced ovarian cancer tumor cell killing in vitro. Significant prolongation of survival by Ad.RGD.pK7.mda-7 in murine ovarian cancer models demonstrates the high clinical translational potential of these viruses for ovarian cancer therapy.


Subject(s)
Adenocarcinoma/therapy , Genetic Therapy/methods , Interleukins/genetics , Ovarian Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/virology , Adenoviridae/genetics , Adenoviridae/pathogenicity , Animals , Cell Line, Tumor , Female , Genetic Vectors/genetics , Humans , Mice , Mice, SCID , Oligopeptides/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/virology , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
5.
Cancer Biother Radiopharm ; 20(5): 467-78, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16248762

ABSTRACT

PURPOSE: This trial was designed to evaluate strategies to improve the efficacy of a radiolabeled monoclonal antibody (mCC49) against tumor-associated glycoprotein-72 (TAG-72) in patients with non-small cell lung cancer (NSCLC). The aims of this study were to determine: safety and maximum tolerated dose (MTD) of (90)Y-mCC49 in combination with interferon alpha2beta (IFN); whether calcium disodium versonate (EDTA) or diethylenetriamine penta-acetic acid (DTPA) could reduce myelosuppression; and safety and MTD of paclitaxel (Taxol) in combination with (90)Y-mCC49. EXPERIMENTAL DESIGN: Patients with advanced (TAG-72 positive) non-small cell lung cancer were entered in three phases; the first was the dose escalation of a single agent (90)Y-mCC49. In the second phase, the dose escalation of (90)Y-mCC49 was attempted with concurrent EDTA or DTPA chelator therapy. In the third phase, radiosensitization with a continuous infusion of paclitaxel (96-hour) was administered with (90)Y-mCC49. All patients received IFN for TAG-72 up-regulation. RESULTS: Thirty-four patients were evaluable. Reversible Grade 4 neutropenia and thrombocytopenia were the dose-limiting toxicities (DLTs). The MTD of (90)Y-mCC49/IFN was 14 mCi/m(2). EDTA did not alter toxicity, while there was a modest reduction of myelosuppression with DTPA. The MTD of continuous infusion paclitaxel in combination with 14 mCi/m(2) of (90)Y-CC49 was 60 mg/m(2). There were no objective tumor responses. CONCLUSIONS: (90)Y-mCC49/IFN was well tolerated at a dose of 14 mCi/m(2). The clinical effect of adjunctive chelating therapy with DTPA was modest. The MTD of coadministered continuous infusion (96-hour) paclitaxel was 60 mg/m(2). Because of the immunogenicity of the murine compound, future studies are planned using a humanized version of CC49.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Chelating Agents/administration & dosage , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Yttrium Radioisotopes/therapeutic use , Adult , Aged , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm/chemistry , Chelating Agents/pharmacology , Clinical Trials as Topic , Disease Progression , Edetic Acid/pharmacology , Female , Glycoproteins/chemistry , Humans , Lung Neoplasms/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Neutropenia , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pentetic Acid/pharmacology , Radioimmunotherapy , Radiometry , Thrombocytopenia , Treatment Outcome , Up-Regulation
6.
J Clin Oncol ; 23(27): 6664-73, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16170174

ABSTRACT

PURPOSE: This is a phase I/IIa study to assess tolerance of gemcitabine and paclitaxel with radiotherapy in locally advanced non-small-cell lung cancer after induction chemotherapy. PATIENTS AND METHODS: Fifty-seven patients with stage III non-small-cell lung cancer were treated with cisplatin 80 mg/m2 on days 1 and 22 and gemcitabine 1,250 mg/m2 on days 1, 8, 22, and 28. Chemoradiotherapy began on day 43 as follows: cohort 1 (n = 9), gemcitabine 300 mg/m2 and paclitaxel 35 mg/m2 weekly (except week 9); cohort 2 (n = 9), gemcitabine 150 mg/m2 and paclitaxel 35 mg/m2 weekly (except week 9); cohort 3 (n = 10) and the 25 phase IIa patients, gemcitabine 300 mg/m2 and paclitaxel 135 mg/m2 every 21 days. Patients were treated with three-dimensional thoracic radiotherapy concurrently to 60 Gy. RESULTS: Weekly chemotherapy resulted in grade 4 esophageal and grade 3 or higher pulmonary toxicities. Reduction in dose density (cohort 3) led to a tolerable toxicity profile and was chosen as the phase IIa regimen. The response rate to induction was 49%, with stable disease in 40% of the patients. The response rate after consolidation therapy was 75% (94% for weekly chemotherapy v 82% for every 3 weeks). Median survival was 23 months, and 3-year survival was 45% for eligible patients. Local relapse occurred in 20% of the patients. Performance status of more than 1 predicted for poor outcome, but baseline pulmonary function did not. Dosimetric parameters including V15, V20, V30 (percent lung volume receiving > or = 15, > or = 20, and > or = 30 Gy, respectively), and mean lung dose correlated with pulmonary toxicity. CONCLUSION: Additional investigation with the 3-week schedule is warranted in patients with a good performance status based on the safety profile and preliminary efficacy data observed in this study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Neoplasm Invasiveness/pathology , Adult , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Cisplatin/therapeutic use , Combined Modality Therapy , Confidence Intervals , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Lung Neoplasms/mortality , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Paclitaxel/therapeutic use , Probability , Radiotherapy, Adjuvant , Remission Induction , Survival Analysis , Gemcitabine
7.
Mol Cancer ; 3: 30, 2004 Oct 13.
Article in English | MEDLINE | ID: mdl-15482594

ABSTRACT

To examine the relationship between mitochondrial DNA (mtDNA) alterations and colorectal tumorigenesis, we used high-resolution restriction endonucleases and sequencing to assess the mitochondrial genome from three histologic subtypes of colorectal adenomas (tubular = 8; tubulovillous = 9; and villous = 8), colorectal cancer (CRC) tissues = 27, and their matched surrounding normal tissue (MSNT) = 52. The mitochondrial genomes were amplified using 9 pairs of overlapping primers and systematically analyzed by means of high-resolution analysis. DNA fragments showing a shift in banding patterns between the three adenomas, CRC, in comparison to the MSNT were sequenced to identify the mtDNA alterations. A total of thirty-eight germ-line mtDNA variants were observed in this study. Twenty-two of the thirty-eight were identified as mutations and 59% (13 of 22) were silent mutations and one was a 1-bp insertion. Sixteen of thirty-eight were distinct SNPs in flanking regions of the restriction sites and, 6 of the 16 (37%) SNPs were not previously reported. Most of these mutations/SNPs were homoplasmic and distributed in various regions of mitochondrial genes including the 16S and 12S rRNA. Based on our results, mtDNA germline variants increased in prevalence with adenoma CRC progression. To the best of our knowledge, this is the first report to show an increased prevalence of mitochondrial gene variants in CRC tumorigenesis.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA, Mitochondrial/genetics , Aged , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , Disease Progression , Genetic Variation , Humans
8.
Gynecol Oncol ; 94(2): 352-62, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15297172

ABSTRACT

OBJECTIVE: Melanoma differentiation associated gene-7 [mda-7/Interleukin (IL)-24] has been identified as a novel anti-cancer agent, which specifically induces apoptosis in cancer cells but not in normal epithelial, endothelial and fibroblast cells. The objective of this study was to evaluate the anti-tumor effect of adenovirus-mediated mda-7/IL-24 (Ad.mda-7) gene therapy in ovarian carcinoma and further improve anti-tumor effect by enhancing infectivity of Ad.mda-7. METHODS: A panel of human ovarian carcinoma cells, OV-4, HEY, SKOV3, SKOV3.ip1 and control normal human mesothelial cells, were infected by a replication deficient recombinant adenovirus encoding mda-7/IL-24 and control virus Ad.CMV.Luc. After 72 h, apoptosis was evaluated by TUNEL and Hoechst staining and further quantified by fluorescent activated cell sorter (FACS) analysis. Infectivity of Ad.mda-7 was enhanced by retargeting it to CD40 or EGF receptors overexpressed on ovarian cancer cells. Subsequently, enhancement in apoptosis of CD40- or epidermal growth factor receptor (EGFR)-retargeted Ad.mda-7 was evaluated. RESULTS: Adenoviral-mediated delivery of mda-7 induces apoptosis ranging from 10-23% in human ovarian cancer cells tested with the highest percentage of apoptosis noted in SKOV3 cells. Minimal apoptosis was noted in normal mesothelial cells. CD40- or EGFR-retargeted Ad.mda-7 increased apoptosis by 10-32% when compared to that achieved with untargeted Ad.mda-7. CONCLUSION: Ad.mda-7 exhibits ovarian cancer-specific apoptosis, but does not affect normal human mesothelial cells. Infectivity enhanced CD40- and EGFR-retargeted Ad.mda-7 augments apoptosis induction, thus increasing the therapeutic index and translational potential of Ad.mda-7 gene therapy.


Subject(s)
Genetic Therapy/methods , Interleukins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/pathogenicity , Adenoviruses, Human/physiology , Apoptosis/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelial Cells/virology , ErbB Receptors/metabolism , Female , Genes, Tumor Suppressor , Humans , Interleukins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/virology , Virus Replication
9.
Gynecol Oncol ; 91(3): 470-5, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14675664

ABSTRACT

OBJECTIVE: Our aim was to determine the outcomes of Stage I uterine papillary serous carcinoma (UPSC) patients with and without adjuvant therapy after comprehensive surgical staging. METHODS: Patients with FIGO Stage I UPSC diagnosed from 1987 to 2000 were identified from tumor registry databases at four institutions. A retrospective chart review identified 60 women who underwent comprehensive surgical staging, including a total hysterectomy, bilateral salpingo-oophorectomy, pelvic/para-aortic lymphadenectomy, and peritoneal cytology. Fisher's exact, chi(2), and log-rank tests were used for the statistical analyses. RESULTS: Of the 60 Stage I patients, 40 (66%) patients received no adjuvant therapy, 12 (20%) received adjuvant radiation therapy (XRT), 7 (12%) received adjuvant chemotherapy (CHM), and 1 (2%) received both XRT and CHM. There were seven recurrences in the observation group versus two recurrences in the XRT group (17% vs 16%, P = 0.9). No recurrences or deaths were observed in the CHM group. The mean disease-free survival rates for the observation and the XRT groups were 31 and 41 months, respectively. The mean overall survival rates for the observation and XRT groups were 39 and 40 months, respectively. The 5-year disease-free survival rates for the observation and XRT groups were 65 and 60%, respectively; the 5-year overall survival rates for observation and XRT groups were 66 and 59%. There was no statistical difference in overall survival among the three groups. CONCLUSION: In this largest reported series of surgical Stage I UPSC patients, recurrence rates were lower than those published in previous studies, suggesting a potential benefit of comprehensive surgical staging in these patients. The risk of recurrence and the mean overall survival were similar between surgical Stage I UPSC patients who were managed conservatively and those treated with adjuvant radiation therapy. These data question the benefit of radiation therapy in UPSC patients with disease confined to the uterus. Finally, given the absence of recurrences and disease-related deaths for adjuvant chemotherapy in these patients, a Phase II/III trial evaluating adjuvant chemotherapy in surgical Stage I UPSC patients should be considered.


Subject(s)
Cystadenocarcinoma, Papillary/therapy , Uterine Neoplasms/therapy , Aged , Aged, 80 and over , Chemotherapy, Adjuvant , Cohort Studies , Cystadenocarcinoma, Papillary/pathology , Cystadenocarcinoma, Papillary/surgery , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Staging , Radiotherapy, Adjuvant , Retrospective Studies , Treatment Outcome , Uterine Neoplasms/pathology , Uterine Neoplasms/surgery
10.
Lung Cancer ; 42(2): 237-45, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14568692

ABSTRACT

Activation of coagulation appears to play a role in tumor progression. This report describes the preliminary results of a phase II study using docetaxel plus enoxaparin in 15 patients with stage IV non-small cell lung cancer (NSCLC). Time to progression was the primary endpoint. Several surrogate markers of coagulation and angiogenesis were evaluated. Enoxaparin was administered at a daily dose of 1 mg/kg (subcutaneously). The initial dose of docetaxel was 100 mg/m2, given as a 60 min infusion every 21 days with prophylactic dexamethasone. Eight patients achieved an objective response (53%) and four had stable disease, with a median duration of 3.5 months. The median time to progression was 5 months (range, 2 to >15 months). The median survival was 11 months. The most frequent toxicities were neutropenia and asthenia. No significant bleeding or thrombotic events were observed. Eleven patients had elevated D-dimer plasma levels prior to therapy, and seven of these patients with a response or stable disease had a significant decline of the D-dimer during therapy. There were no consistent changes of the plasma levels of the angiogenic factors, except for transforming growth factor-beta-1 (TGF-beta1). The median baseline level of TGF-beta1 prior to therapy was 34,867 pg/ml. Twelve out of 13 patients who achieved a response or stable disease had a significant reduction of the TGF-beta1 levels during therapy. Enoxaparin in combination with chemotherapy was safe and well tolerated in patients with advanced NSCLC. This preliminary data suggests that enoxaparin may prolong the time to progression, and therefore justify the continuation of this trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Aged , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Docetaxel , Enoxaparin/administration & dosage , Enoxaparin/adverse effects , Female , Humans , Infusions, Intravenous , Injections, Subcutaneous , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , Taxoids/administration & dosage , Taxoids/adverse effects , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta1
11.
J Gene Med ; 5(10): 839-851, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14533192

ABSTRACT

BACKGROUND: Systemic adenoviral (Ad) gene therapy for renal disorders is largely hampered by the unique architecture of the kidney. Consequently, currently available Ad vectors are of only limited therapeutic utility in the context of glomerular and fibroproliferative renal diseases. METHODS: The Ad vectors studied in the context of blocking renal fibrosis were AdTbeta-ExR and AdCATbeta-TR. AdTbeta-ExR encodes a chimeric soluble molecule comprising the entire ectodomain of the human type II TGF-beta receptor, genetically fused to the Fc fragment of the human IgG1 (sTbetaRII), while AdCATbeta-TR encodes only the dominant-negative truncated ectodomain of the human type II TGF-beta receptor. The biologic activity of the type II TGF-beta receptor was evaluated in vitro by its ability to inhibit cellular proliferation and in vivo in a unilateral ureter obstruction fibrosis model. Renal targeting with sTbetaRII was evaluated immunohistochemically after intramuscular (IM) delivery of AdTbeta-ExR. The renal antifibrotic effect of the Ad vectors was evaluated in a lupus murine model with both light and electron microscopy and urinalysis. RESULTS: sTbetaRII was detected in the glomeruli after remote IM injection of AdTbeta-ExR, but not the control AdCATbeta-TR, indicating renal deposition of the heterologous soluble fusion protein after its expression in the muscle and secretion into the circulation. AdTbeta-ExR, but not AdCATbeta-TR, could transiently inhibit mesangial expansion, glomerular hypercellularity, proteinuria and cortical interstitial fibrosis in a murine lupus model. However, the autoimmune renal disease eventually surpassed the antifibrotic effect. CONCLUSIONS: These results indicate the superiority of a soluble type II TGF-beta receptor over a dominant-negative, non-soluble type II TGF-beta receptor in the context of blocking renal fibrosis in murine models.


Subject(s)
Genetic Therapy , Kidney Diseases/therapy , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Adenoviridae/genetics , Animals , Disease Models, Animal , Fibrosis/therapy , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Glomerulonephritis/genetics , Glomerulonephritis/therapy , Injections, Intramuscular , Kidney/pathology , Kidney Diseases/genetics , Mice , Mice, Inbred BALB C , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Recombinant Fusion Proteins/genetics
12.
Cancer Biother Radiopharm ; 18(3): 393-404, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12954125

ABSTRACT

CC49 is a second-generation murine antibody with anti-TAG-72 (tumor-associated antigen) reactivity. For cancer therapy, it has the advantage of being expressed on adenocarcinomas but not on most normal tissues. CC49 has been utilized in phase I and II clinical trials at multiple institutions. Therapeutic applications to date have included (131)I-, (90)Y-, and (177)Lu-CC49, with tracer amounts of (111)In-CC49 as a dosimetry surrogate for (90)Y-CC49 therapy. Dosimetry methods and details of their description vary between studies. Biodistribution to normal organs and the effective plasma T(1/2) for various radionuclides were relatively consistent among patients with different diseases and treatment at several institutions. As expected with marrow suppression being the dose-limiting toxicity, higher doses of (177)Lu-CC49 were tolerated via intraperitoneal than IV administration. The biologic response modifier interferon enhanced TAG-72 expression and resulted in a trend of increased uptake of (131)I-CC49 by tumors. Tumor dose estimates were more variable than that of normal organs. Standardization and improved dosimetry may be helpful for comparison among patients in various studies and for establishing dose/toxicity relationships that are useful for predicting safe levels of radioimmunoconjugates.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neoplasm/immunology , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Neoplasms/metabolism , Radioisotopes/administration & dosage , Radioisotopes/pharmacokinetics , Clinical Trials as Topic , Half-Life , Humans , Radiometry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Treatment Outcome
13.
Clin Cancer Res ; 9(7): 2701-10, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855650

ABSTRACT

PURPOSE: Vascular endothelial growth factor (VEGF) is a potent angiogenic agent and plays a major role in tumor growth and metastases. We have previously reported the locoregional (i.p.) delivery of adenovirus-mediated antiangiogenic soluble FLT-1 (sFLT-1; a naturally encoded potent VEGF antagonist) gene therapy to inhibit VEGF action in a murine ovarian carcinoma model. This study was predicated on the fact that systemic delivery of sFLT-1 might allow an approach for therapy of disseminated tumor. The purpose of this study is to test the effects of i.v. delivered, adenovirus-mediated sFLT-1 on the survival duration in a murine ovarian tumor model and to evaluate the safety of i.v.-delivered versus i.p.-delivered adenovirus-mediated sFLT-1 in non-tumor-bearing mice. EXPERIMENTAL DESIGN: To determine the effects of i.v.-administered adenovirus-mediated sFLT-1 on survival duration of mice bearing i.p. human ovarian tumors, an E1A/B-deleted, (replication-deficient) infectivity-enhanced recombinant adenovirus AdRGDGFPsFLT-1 encoding cDNA for both sFLT-1 and GFP (green fluorescent protein), a control adenovirus AdRGDGFP encoding GFP alone, or PBS was delivered i.v. The therapeutic effect of sFLT-1 was evaluated by survival duration of the mice. Furthermore, the safety of i.v.- or i.p.-delivered adenovirus-mediated sFLT-1 was evaluated by administering AdRGDGFPsFLT-1, AdRGDGFP, or PBS either i.v. or i.p. into non-tumor-bearing mice. Adenovirus-mediated gene expression was determined by determining GFP expression using fluorescent microscopy and by assessing sFLT-1 expression in liver, lungs, spleen, and kidneys by immunohistochemistry using anti-FLT-1 monoclonal antibody. Systemic levels of sFLT-1 were evaluated by ELISA and the toxicity was evaluated by histopathology. RESULTS: The i.v. delivery of AdRGDGFPsFLT-1 in the ovarian tumor model resulted in a shorter duration of survival of the mice as compared with the control group. Furthermore, in the safety evaluation experiment, i.v. administration of AdRGDGFPsFLT-1 in non-tumor-bearing mice principally localized to the liver. This localization lead to sFLT-1 overexpression, mainly in the liver, resulting in hemorrhage and tissue toxicity. However, i.p. delivery of AdRGDGFPsFLT-1 did not localize principally to the liver, leading to negligible expression of sFLT-1, and no intrahepatic hemorrhage or toxicity was observed. The i.v. delivery of the control virus AdRGDGFP also principally localized to the liver, leading to GFP expression mainly in the liver. However, neither hemorrhage nor morphological cytotoxicity was observed. i.p. delivery of AdRGDGFP resulted in ectopic localization to the liver with very little GFP expression and no toxicity. These results suggest that overexpression of sFLT-1 in the liver as a result of i.v. delivery is hepatotoxic. CONCLUSIONS: Our results suggest that i.v. delivery of the sFLT-1 gene via replication-deficient, infectivity-enhanced recombinant adenoviral vectors will result in overexpression of sFLT-1 in the liver leading to unacceptable hepatotoxicity. Tumor-specific targeting of the vectors and tumor-specific expression strategies should be used to ensure a clinically useful antiangiogenesis gene therapy.


Subject(s)
Adenoviridae/genetics , Extracellular Matrix Proteins/therapeutic use , Liver/drug effects , Animals , Cell Division , Cell Line, Tumor , Cell Survival , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins/toxicity , Female , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins , Humans , Immunohistochemistry , Liver/metabolism , Luminescent Proteins/metabolism , Mice , Mice, SCID , Microscopy, Fluorescence , Myosin Heavy Chains , Neoplasm Metastasis , Neovascularization, Pathologic , Nonmuscle Myosin Type IIB , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Time Factors , Vascular Endothelial Growth Factor Receptor-1
14.
Biol Blood Marrow Transplant ; 8(11): 619-24, 2002.
Article in English | MEDLINE | ID: mdl-12463481

ABSTRACT

High-dose busulfan is widely used in allogeneic and autologous marrow transplantation preparative regimens. Variation in the area under the concentration/time curve (AUC) for oral busulfan results in substantial risk of over or under treatment with excess risk of toxicity or relapse. Use of the IV formulation reduces this variability by eliminating variability in absorption. Variability due to drug metabolism remains, but simplified pharmacokinetic study may be employed to achieve a specific target AUC. In conventional sampling strategies for determining AUC after oral administration, 12 samples are used over 6 hours to assure accurate tracking of erratic absorption. With IV busulfan there is no necessity for measuring plasma levels during the infusion because busulfan pharmacokinetics are well described with a single-compartment, first-order elimination model. In theory, only peak and trough levels should be necessary, but for assurance of reliability in clinical decision making, it must be possible to identify outlier values. This process requires at least 4 samples. We studied a total of 59 adult patients receiving a 2-hour IV busulfan infusion to develop a limited sampling strategy (LSS). At the end of a 2-hour infusion, we collected 11 samples from 18 patients and compared the AUC obtained when all samples were used with the AUC obtained when samples were collected only hourly. The mean AUC calculation was 5% higher (1002 versus 956 microM-min) and the coefficient of variation (CV) was substantially better (4.6% versus 8.2%) when only the postinfusion samples were used. A follow-up study of 41 consecutive patients demonstrated that all patients were easily evaluable with a coefficient of variation (CV) for the AUC of 2.6%. To validate this approach, we analyzed pharmacokinetic data on 60 patients in the phase II clinical trial of the IV formulation described by Anderson et al. Data on an additional 36 patients from a companion study also were analyzed. The AUC based on all 11 samples from each patient were compared with the AUC based on the 5 postinfusion samples. The results of this analysis confirmed comparable reliability and possibly superior precision of the University of Alabama at Birmingham 5-sample LSS. These results validated that LSS for IV busulfan will make possible meaningful and accurate comparisons of busulfan versus TBI-based preparative regimens and comparison of dose intensity of busulfan-containing preparative regimens in trials of submyeloablative transplantation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Busulfan/pharmacokinetics , Drug Monitoring/methods , Antineoplastic Agents, Alkylating/administration & dosage , Area Under Curve , Busulfan/administration & dosage , Clinical Trials, Phase II as Topic , Drug Monitoring/statistics & numerical data , Half-Life , Hematologic Neoplasms/drug therapy , Humans , Infusions, Intravenous , Models, Statistical , Reproducibility of Results
15.
Clin Cancer Res ; 8(10): 3092-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12374676

ABSTRACT

PURPOSE: Our purpose in the study was to establish the maximum tolerated dose and toxicity profile of SGN-10 (or BR96 sFv-PE40), a single-chain immunotoxin. SGN-10 is composed of the fused gene products encoding the translocating and ADP-ribosylating domains of Pseudomonas exotoxin (PE40) and the variable heavy (V(H)) and variable light (V(L)) regions of BR96 monoclonal antibody. This antibody is specific for a Lewis(Y) (Le(Y))-related carbohydrate antigen expressed on multiple carcinomas. EXPERIMENTAL DESIGN: A total of 46 patients with Le(Y)-positive metastatic carcinoma were enrolled in a Phase I dose-escalation study in cohorts of three to six patients who received SGN-10 at doses ranging from 0.024 to 0.962 mg/m(2), administered on days 1, 4, 8, and 11, followed by 2 weeks of rest and a second cycle of therapy. Pharmacokinetics and human antibody response to SGN-10 were also determined. RESULTS: The maximum tolerated dose of SGN-10 was 0.641 mg/m(2) with gastrointestinal dose-limiting toxicity. Pharmacokinetic studies performed in eight patients at the 0.641-mg/m(2) dose revealed a t([1/2]) of 2.5 +/- 0.3 h and a C(max) of 389 +/- 112 ng/ml. Pharmacodynamic analyses demonstrated a rapid clearance of the drug by day 11 associated with an antitoxin human antitoxin antibody (HATA) response in most patients. Signs consistent with a modest vascular leak syndrome, specifically, transient hypoalbuminemia, were observed in patients treated with doses of > or =0.384 mg/m(2). No complete or partial tumor responses were observed at an 8-week evaluation, although 31% of patients had stable disease. CONCLUSIONS: The maximal tolerated dose of SGN-10 given twice weekly for 2 weeks is 0.641 mg/m(2) with gastrointestinal dose-limiting toxicity. The immunogenicity of the toxin moiety limits the ability of SGN-10 to circulate by day 11 of therapy. Studies are ongoing to evaluate strategies to ameliorate toxicities and to inhibit the development of the anti-SGN-10 immune response.


Subject(s)
Immunotoxins/therapeutic use , Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Adult , Aged , Antibodies, Monoclonal , Cohort Studies , Female , Humans , Immunotoxins/adverse effects , Immunotoxins/pharmacokinetics , Lewis Blood Group Antigens/immunology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
16.
Clin Cancer Res ; 8(9): 2806-11, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231520

ABSTRACT

PURPOSE: The purpose of this study was to determine the feasibility and maximum tolerated dose of (90)Yttrium-CC49 ((90)Y-CC49) as the radioimmunotherapy (RIT) component of an i.p. combined modality treatment for recurrent ovarian cancer. EXPERIMENTAL DESIGN: A Phase I trial of (90)Y-CC49 RIT was conducted in ovarian cancer patients who had persistent or recurrent intra-abdominal disease, had failed one or two prior chemotherapy regimens, and demonstrated TAG-72 expression. Patients were treated with a previously established combined modality treatment protocol of s.c. IFN alpha2b, i.p. paclitaxel, and increasing dosages of i.p. (90)Y-CC49. Patients were monitored for toxicity, generation of human antimouse antibody response, and clinical efficacy. RESULTS: Twenty eligible patients were treated per study specifications. All patients had been treated with debulking and paclitaxel/carboplatin-based chemotherapy at initial diagnosis. The patients included 11 patients with persistent disease at the time of second look laparotomy and 9 patients with delayed recurrence. Patients were treated with i.p. (90)Y-CC49 given in combination with s.c. IFN alpha2b (dose of 3 x 10(6) units for a total of four doses) and i.p. paclitaxel (dose of 100 mg/m(2)). RIT treatment was associated with primarily hematological toxicity. The maximum tolerated dose of i.p. (90)Y-CC49 was established at 24.2 mCi/m(2) in this combined regimen. Of nine patients with measurable disease, two had partial responses lasting 2 and 4 months. Of 11 patients with nonmeasurable disease, median time to progression was 6 months in 7 patients who recurred; 4 of these patients remain no evidence of disease at 9+, 18+, 19+, and 23+ months. CONCLUSIONS: (90)Yttrium-CC49-based RIT in combination with IFN alpha2b and i.p. paclitaxel is feasible and well tolerated at a dose of < or =24.2 mCi/m(2).


Subject(s)
Adenocarcinoma/radiotherapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Immunoconjugates/therapeutic use , Ovarian Neoplasms/radiotherapy , Radioimmunotherapy , Yttrium Radioisotopes/therapeutic use , Adult , Aged , Animals , Antibodies, Heterophile/biosynthesis , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/immunology , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Immunoconjugates/administration & dosage , Injections, Intraperitoneal , Maximum Tolerated Dose , Mice , Middle Aged , Treatment Outcome , Yttrium Radioisotopes/administration & dosage
17.
Cancer Res ; 62(15): 4273-81, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12154029

ABSTRACT

Metastatic renal cell carcinoma (RCC) is one of the most treatment-resistant malignancies in humans. Therefore, the identification of new agents with better antitumor activity merits a high priority in the treatment of advanced RCC. In this regard, gene therapy with adenoviral (Ad) vectors is a promising new modality for cancer. However, a primary limiting factor for the use of Ad vectors for cancer gene therapy is their critical dependence on cellular expression of the primary Ad receptor, the coxsackie and adenovirus receptor (CAR), known to be down-regulated in many cancer types. Following the identification of CAR deficiency in RCC lines, we have found abundant membrane expression of alpha(v)beta 3 and alpha(v)beta 5 integrins and of the putative receptor to Ad serotype 3 (Ad3). As an alternative gene therapy approach for RCC that would circumvent CAR deficiency, we employed retargeting of replication-incompetent Ad vectors and replication-competent Ad viruses to alpha(v)beta 3 and alpha(v)beta 5 integrins and to the putative Ad3 receptor. These strategies to genetically alter Ad tropism were based on either the insertion of a cysteine-aspartate-cysteine-arginine-glycine-aspartate-cysteine-phenylalanine-cysteine (RGD) motif into the HI loop of the Ad fiber knob domain or on generation of a chimeric Ad fiber composed of adenovirus serotype 5 shaft/Ad3 knob. Both strategies proved highly efficient to circumvent CAR deficiency and enhance gene delivery into RCC cells. Furthermore, in the context of replication-competent Ad, tropism alteration resulted in distinct capacity of the retargeted viruses to infect, replicate, and lyse RCC models in vitro and in vivo. The retargeting strategies were particularly beneficial in the context of replication-competent Ad. These findings underscore the importance of CAR-independent cellular entry mechanisms in RCC and are highly consequential for the development of viral antitumor agents for RCC and other CAR-negative tumors.


Subject(s)
Adenoviridae/genetics , Carcinoma, Renal Cell/therapy , Genetic Therapy/methods , Integrins/metabolism , Kidney Neoplasms/therapy , Receptors, Vitronectin/metabolism , Adenoviridae/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/virology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/virology , Mice , Mice, Nude , Receptors, Virus/deficiency , Receptors, Virus/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Lung Cancer ; 37(2): 189-99, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12140142

ABSTRACT

The purpose of this trial was to evaluate the feasibility of concurrent paclitaxel/cisplatin and conventional thoracic irradiation in locally advanced non-small cell lung cancer (NSCLC). Ambulatory patients with medically inoperable or unresectable stage II-III NSCLC, and performance status 0-2 were eligible. Patients were not excluded from this trial if they had lost more than 5% of their body weight during the preceding 3 months, and/or if they had small ipsilateral pleural effusion. The initial dose of paclitaxel/cisplatin was 110 and 50 mg/m(2), and was escalated through five dose levels. Four cycles of chemotherapy were planned; the first two cycles were given concurrently with radiotherapy (4 weeks apart), followed by two additional cycles (every 3 weeks). Conventional chest radiotherapy to a total dose of 60 Gy (2 Gy per day) was delivered in 6 weeks. Forty-three patients were enrolled of which 38 were evaluable for response. Dose-limiting toxicities were grade 4 neutropenia (43% of patients) and grade 3 esophagitis (26% of patients) during the chemoradiotherapy phase. Grade > or = 2 acute and late pulmonary toxicity occurred in 10 and 68% of the patients, respectively. In most patients, prompt symptomatic and radiologic improvement was observed with early steroid administration. The volume of lung receiving 15-30 Gy was correlated with late pulmonary toxicity. The overall response was 84% with ten complete and 22 partial responses. The median survival was 16.5 months (95% confidence interval, 9.5 to 25) for those patients evaluable for response. After a median follow-up of 70 months, 5 (13%) patients are alive without evidence of disease. The maximum tolerated dose (MTD) of paclitaxel and cisplatin with concurrent radiotherapy is at dose level 3 paclitaxel (135 mg/m(2)) and cisplatin (75 mg/m(2)). Toxicity, although significant, was manageable in the great majority of the patients. The activity observed with this regimen is particularly noteworthy when considering the advanced nature of these patients, and the fact that patients (N=18) with poor risk factors were included in the study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Adolescent , Adult , Cisplatin/administration & dosage , Combined Modality Therapy , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Respiratory Function Tests , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL