Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 403
Filter
1.
Ann Oncol ; 26(11): 2267-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26410620

ABSTRACT

BACKGROUND: The efficacy and safety of nab-paclitaxel versus dacarbazine in patients with metastatic melanoma was evaluated in a phase III randomized, controlled trial. PATIENTS AND METHODS: Chemotherapy-naïve patients with stage IV melanoma received nab-paclitaxel 150 mg/m(2) on days 1, 8, and 15 every 4 weeks or dacarbazine 1000 mg/m(2) every 3 weeks. The primary end point was progression-free survival (PFS) by independent radiologic review; the secondary end point was overall survival (OS). RESULTS: A total of 529 patients were randomized to nab-paclitaxel (n = 264) or dacarbazine (n = 265). Baseline characteristics were well balanced. The majority of patients were men (66%), had an Eastern Cooperative Oncology Group status of 0 (71%), and had M1c stage disease (65%). The median PFS (primary end point) was 4.8 months with nab-paclitaxel and 2.5 months with dacarbazine [hazard ratio (HR), 0.792; 95.1% confidence interval (CI) 0.631-0.992; P = 0.044]. The median OS was 12.6 months with nab-paclitaxel and 10.5 months with dacarbazine (HR, 0.897; 95.1% CI 0.738-1.089; P = 0.271). Independently assessed overall response rate was 15% versus 11% (P = 0.239), and disease control rate (DCR) was 39% versus 27% (P = 0.004) for nab-paclitaxel versus dacarbazine, respectively. The most common grade ≥3 treatment-related adverse events were neuropathy (nab-paclitaxel, 25% versus dacarbazine, 0%; P < 0.001), and neutropenia (nab-paclitaxel, 20% versus dacarbazine, 10%; P = 0.004). There was no correlation between secreted protein acidic and rich in cysteine (SPARC) status and PFS in either treatment arm. CONCLUSIONS: nab-Paclitaxel significantly improved PFS and DCR compared with dacarbazine, with a manageable safety profile.


Subject(s)
Albumins/therapeutic use , Dacarbazine/therapeutic use , Melanoma/diagnosis , Melanoma/drug therapy , Paclitaxel/therapeutic use , Skin Neoplasms/diagnosis , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/therapeutic use , Disease-Free Survival , Female , Humans , Male , Middle Aged , Young Adult
2.
Cancer Immunol Immunother ; 50(8): 397-407, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11726134

ABSTRACT

The epithelial mucin MUC1 is considered an opportune target antigen for cancer immunotherapy, as it is over-expressed and exhibits aberrant glycosylation in malignant cells. Because dendritic cells (DC) are powerful initiators of immune responses, efforts have focused on tumor antigen-bearing DC as potent cancer vaccines. In this study we have characterized the transduction of monocyte-derived DC with a highly attenuated vaccinia virus vector [modified vaccinia Ankara (MVA)] encoding human MUC1 and the immunostimulatory cytokine IL-2. Analysis of transduced DC cultures generated from a number of donors revealed MUC1 expression in the range of 27-54% of the cells and a co-regulated secretion of bioactive IL-2. As shown by FACS analysis with MUCI-specific antibodies, the MVA-MUC1/IL-2-transduced DC predominantly expressed the fully processed glycoform of MUC1, typical of that displayed by normal epithelia. Over a 3-day period after transduction, transgene expression declined concurrent with an increase in MVA-induced cytopathic effects. The transduced DC stimulated allogeneic lymphocyte proliferation, indicating that DC immunostimulatory function is not impaired by vector transduction. In the presence of IL-2, MVA-transduced DC were able to enhance autologous lymphocyte proliferation. Also, vector expression was analyzed in DC cultures treated with TNF-alpha, a known DC maturation factor. As indicated by the up-regulation of several DC maturation markers, neither virus infection nor transgene expression influenced the maturation capacity of the cells. The MVA-MUC1/IL-2 vector effectively transduced both immature and TNF-alpha-matured DC. Overall, our results are encouraging for the clinical application of MVA-MUC1/IL-2-transduced DC.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Genetic Therapy , Interleukin-2/immunology , Mucin-1/immunology , Antigens, Neoplasm/genetics , Apoptosis , Blood Donors , Cells, Cultured/metabolism , Dendritic Cells/metabolism , Dose-Response Relationship, Immunologic , Genetic Vectors/genetics , Glycosylation , Humans , Interleukin-2/genetics , Kinetics , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mucin-1/genetics , Mucin-1/metabolism , Protein Processing, Post-Translational , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Transfection , Transgenes , Tumor Necrosis Factor-alpha/pharmacology , Vaccinia virus/genetics
3.
Clin Cancer Res ; 7(8): 2285-91, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489803

ABSTRACT

Cutaneous melanoma is one of the most rapidly increasing cancers in the United States. Because of the lack of effective treatment options and toxicities of most chemotherapeutic and radiation regimes, immunotherapies such as vaccination therapy represent an attractive approach for patients with advanced melanoma. The purpose of this study was to evaluate the response rate, time to progression, and survival of patients with metastatic melanoma treated by direct intratumoral injection with Allovectin-7 (a plasmid DNA encoding the genes HLA-B7 and beta2-microglobulin complexed with a cationic lipid mixture, DMRIE/DOPE. Fifty-two patients with metastatic melanoma were enrolled in this Phase II study. Therapy consisted of six intratumoral injections of 10 microg of Allovectin-7 over a 9-week period. Treatment was well tolerated. Treatment-related adverse events were mild to moderate, the most frequent of which were ecchymosis, pruritus (and/or discomfort at the injection site), and pneumothoraces. Regression of the injected lesion was observed in 18% of patients, including one complete response, three partial responses, and five minor responses. An overall response rate of 4% (two partial responses) was documented, and nine patients (18%) maintained stable disease for at least 11 weeks. Six patients remained alive 25.1 to 39.4 months from their first injection, including two patients with local (injected tumor) responses and one patient with an overall disease partial response. This study demonstrates that intratumoral administration of Allovectin-7 in metastatic melanoma is safe and can produce both responses in injected lesions and in overall disease. Clinical trials optimizing patient selection and combining Allovectin-7 with other modalities of therapy are currently ongoing in an effort to improve response rates.


Subject(s)
DNA , Lipids/therapeutic use , Melanoma/therapy , Plasmids/therapeutic use , Adult , Aged , Aged, 80 and over , DNA, Recombinant , Erythema/chemically induced , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Injections, Intralesional/adverse effects , Lipids/administration & dosage , Lipids/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Pain/etiology , Plasmids/administration & dosage , Plasmids/adverse effects , Pruritus/chemically induced , Survival Analysis , Time Factors , Treatment Outcome
4.
Anticancer Drugs ; 12(3): 213-20, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11290869

ABSTRACT

The azonafides are a series of anthracene-based DNA intercalators which inhibit tumor cell growth in vitro at low nanomolar concentrations and are not affected by the multidrug resistance phenomenon (MDR). Prior studies have described antitumor efficacy in murine tumor models including L-1210 and P-388 leukemias, and B-16 melanoma. The current results extend these cell line observations to human tumors tested in the NCI panel of 56 cell lines, in freshly isolated tumors tested in colony-forming assays in soft agar and in several animal models. In the NCI panel, the overall mean 50% cell kill (LC50) for the unsubstituted azonafide, AMP-1, was 10(-5.53) M, with some selectivity noted in melanomas (10(-6.22) M). The mean LC50 for the 6-ethoxy substituted analog, AMP-53, was 10(-5.53) M, with some selectivity found in non-small cell lung cancer (10(-5.91)) and renal cell carcinoma (10(-5.84)). In freshly isolated human tumors tested in soft agar, there was marked activity (mean IC50 in microg/ml) for AMP-53 in four cell types: breast cancer (0.09), lung cancer (0.06), renal cell carcinomas (0.06) and multiple myeloma (0.03). These effects were superior to doxorubicin and to several other azonafides, including AMP-1, AMP-104 and the 6-hydroxyethoxy derivative, AMP-115. Compound AMP-1 was shown to be superior to amonafide in the mammary 16C breast cancer model in B6CF31 mice, but it had little activity in Colon-38 nor in M5076 ovarian sarcomas in vivo. Nine azonafides were evaluated in the Lewis lung cancer model in C57/bl mice, but only AMP-53 demonstrated significant efficacy with a treated/control x 100% (T/C) value of 30%. Because AMP-53 demonstrated the greatest breadth of activity, it was then evaluated in several human tumor cell lines growing in mice with severe combined immunodeficiency disease (SCID). Only three tumors were sensitive (T/C<42%), including HL-60 leukemia (T/C=39%), MCF-7 breast cancer (T/C=39%) and A549 non-small cell lung cancer (T/C=37%). Overall, these results demonstrate that the 6-ethoxy substituted azonafide, AMP-53, has consistent (in vitro and in vivo) experimental antitumor activity in human breast and lung cancer, and could be considered for clinical testing in patients with MDR tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Imides/pharmacology , Intercalating Agents/pharmacology , Isoquinolines/pharmacology , Tumor Cells, Cultured/drug effects , Adenine , Animals , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Naphthalimides , Neoplasms/drug therapy , Organophosphonates , Tumor Stem Cell Assay
5.
Cancer Chemother Pharmacol ; 47(1): 83-8, 2001.
Article in English | MEDLINE | ID: mdl-11221967

ABSTRACT

PURPOSE: To evaluate the in vitro activity of polyethylene glycol-conjugated L-asparaginase (PEG-Lasparaginase) against fresh human tumor specimens, using the human tumor clonogenic assay (HTCA), and to perform a phase I dose-escalation clinical trial of PEG-L-asparaginase. The goal of the clinical study was to determine the toxicity and optimum biologic dose of PEG-L-asparaginase based on depletion of serum L-asparagine in patients with advanced solid tumors. METHODS: A modified method for determination of serum L-asparagine is described. PEG-L-asparaginase was administered by intramuscular injection every 2 weeks to 28 patients with various types of advanced solid tumor malignancies. At least 3 patients were evaluated at each dose level: 250 IU/m2, 500 IU/m2, 1,000 IU/m2, 1,500 IU/m2, 2,000 IU/m2. RESULTS: The in vitro HTCA studies suggested good antitumor activity against malignant melanoma and multiple myeloma. Serum L-asparagine was most consistently and profoundly depleted (up to 4 weeks) in patients treated with 2,000 IU/m2. Patients receiving this dose level also showed more frequent grade 1, grade 2, and occasional grade 3 toxicities of fatigue/weakness, nausea/vomiting, and anorexia/ weight loss. Three patients developed hypersensitivity reactions, but these were not dose related. Two patients developed deep vein thromboses. We saw no episodes of clinical pancreatitis, but there were minor fluctuations of serum amylase and lipase. We saw no partial or complete responses in patients treated in this study, including 11 patients with malignant melanoma. CONCLUSIONS: We conclude that PEG-L-asparaginase is generally well tolerated in patients with advanced solid tumors, and a dosage of 2,000 IU/m2 by intramuscular injection every 2 weeks results in significant depletion of serum L-asparagine.


Subject(s)
Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Glutaminase/therapeutic use , Neoplasms/drug therapy , Polyethylene Glycols/therapeutic use , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Asparaginase/administration & dosage , Asparaginase/adverse effects , Asparagine/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Screening Assays, Antitumor , Glutaminase/administration & dosage , Glutaminase/adverse effects , Humans , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Neoplasm Proteins/blood , Neoplasms/blood , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Skin Neoplasms/drug therapy
6.
Cell Immunol ; 204(2): 96-104, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11069717

ABSTRACT

Natural killer (NK) cells play an important role in combating infectious and malignant diseases and interleukin-2 (IL-2) has been shown to promote proliferation and activation of NK cells in vitro and in vivo. Here we investigate the effects of local cationic lipid-mediated IL-2 gene transfer on intratumoral accumulation and activation of NK cells in a SCID mouse tumor model. UM449 human melanoma tumors in SCID mice received intratumoral injections of DMRIE/DOPE admixed with VR1103, a DNA plasmid encoding the gene for human IL-2. Dissagregated tumor cells were tested for IL-2 secretion and were characterized using antibodies to asGM1, MAC-1, and F4/80 antigens. Granzyme A, a proteolytic serine esterase, was also measured in tumor cell lysates. IL-2 secretion from tumors injected with VR1103:DMRIE/DOPE peaked at 48 h after injection and fell to baseline levels on day 8. Intratumoral granzyme A activity was significantly increased in tumors injected with IL-2 plasmid:DMRIE/DOPE complexes, but not by an irrelevant plasmid DNA:DMRIE/DOPE control. Importantly, the growth of UM449 tumors was slowed in VR1103:DMRIE/DOPE-injected tumors. These results indicate that local cationic lipid-mediated gene transfer of IL-2 induces activation of intratumoral NK cells and slows tumor growth.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-2/biosynthesis , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Melanoma/immunology , Animals , Cations , Chemotaxis, Leukocyte , Gene Transfer Techniques , Granzymes , Humans , Interleukin-2/genetics , Lipids , Mice , Mice, SCID , Serine Endopeptidases/biosynthesis , Transgenes , Transplantation, Heterologous
7.
Clin Cancer Res ; 6(10): 3904-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11051236

ABSTRACT

Tumor-infiltrating CD8+ T-lymphocytes (T-TILs) are thought to be relevant to immunosurveillance of several tumor types including B-cell non-Hodgkin's lymphoma. B- and T-lymphocyte interactions via cellular adhesion molecules (CAMs), recognition molecules (HLAs), and costimulatory molecules (CSMs) are necessary for optimal antigen-specific T-cell activation to occur and may be important in generating effective host T-TIL responses. We previously found that low T-TIL response (CD8+ T cells < 6%) correlates with statistically shorter relapse-free survival in patients with diffuse large-cell lymphoma (DLCL). We now extend our observations in 71 DLCL patients by analyzing malignant B-cell expression of the following molecules important in T-cell activation: (a) recognition molecules [MHC I (MAS and MCA) and MHC II (HLA-DR, -DP, -DQ)]; (b) CAMs [leukocyte function antigen 1 (CD11a and CD18) and intracellular adhesion molecule 1 (CD54)]; and (c) CSMs [B7.1 (CD80) and B7.2 (CD86)]. Eighteen patients (25%) had low a T-TIL response, and 53 patients (75%) had a high T-TIL response. Overall, expression of the MHC class H molecules HLA-DR and HLA-DQ was most conserved. The loss of B7.2 (P = 0.04), intracellular adhesion molecule 1 (P = 0.0004), MAS (P = 0.02), and HLA-DR (P = 0.0004) expression was significantly associated with decreased T-TIL response. In 100% of patients with low T-TIL responses, at least one HLA, CAM, or CSM was undetectable on the malignant B cells by immunohistochemical staining (mean number of molecules lost = 2.67). In contrast, 49% of patients with high T-TIL responses had no losses in HLA, CAM, or CSM expression (mean number of molecules lost = 0.89). The mean number of absent molecules (HLA, CAM, or CSM) was significantly associated with T-TIL response (P = 0.0001). We conclude that loss of HLA, CAM, or CSM expression on malignant B cells is associated with a poor host T-cell immune response. In addition, because patients with low T-TIL response had lost expression of multiple cellular adhesion, recognition, and costimulatory molecules, our results suggest that a combination of immunorestorative therapies may be required to generate effective antitumor T-cell responses in B-cell DLCL.


Subject(s)
Antigens, CD/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Membrane Glycoproteins/metabolism , B7-1 Antigen/blood , B7-2 Antigen , CD18 Antigens/blood , Cell Adhesion , HLA-DP Antigens/blood , HLA-DQ Antigens/blood , HLA-DR Antigens/blood , Humans , Immunohistochemistry , Lymphocyte Function-Associated Antigen-1/blood , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/pathology
8.
Cancer Gene Ther ; 7(6): 853-60, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10880015

ABSTRACT

Cationic lipid-mediated gene transfer is a safe and effective means of delivering potent immunomodulatory cytokines directly into tumors. This approach avoids undesirable side effects, including systemic toxicities. To investigate key factors affecting intratumoral (i.t.) gene transfer, cationic lipid-DNA complexes were injected into subcutaneous human melanoma tumors in severe combined immunodeficient mice. Animals received i.t. injections of VR1103, a DNA plasmid encoding the gene for human interleukin-2 (IL-2), either alone or complexed with the cationic lipid N-(1-(2,3-dimyristyloxypropyl)-N,N-dimethyl-(2-hydroxyethyl) ammonium bromide/dioleoyl phosphatidylethanolamine (DMRIE/DOPE). Tumors were subcultured and supernatants were tested for IL-2 secretion by enzyme-linked immunosorbent assay. IL-2 secretion was consistently higher when lipid:DNA (L:D) complexes were formulated at high L:D ratios (wt/wt), and IL-2 transgene expression increased in a DNA dose-dependent manner. A comparison of naked plasmid and lipid-complexed DNA revealed that lipid complexes were more effective for i.t. gene transfer. Using an enhanced green fluorescent protein reporter plasmid and flow cytometry, i.t. transfection efficiency was 1.74% (+/- 1.08%). Tumor injection technique, including injection volume and location, had a limited impact on i.t. gene transfer. These results indicate that the formulation and dosage of cationic L:D complexes, but not injection technique, play a key role in determining the level of i.t. transgene expression.


Subject(s)
DNA/metabolism , Glycerophospholipids/metabolism , Lipid Metabolism , Melanoma/genetics , Phosphatidylethanolamines , Plasmids/genetics , Quaternary Ammonium Compounds/metabolism , Skin Neoplasms/genetics , Transfection/methods , Animals , Flow Cytometry , Genetic Therapy , Green Fluorescent Proteins , Humans , Immunotherapy , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lipids , Luminescent Proteins/metabolism , Melanoma/metabolism , Mice , Mice, SCID , Mice, Transgenic , Skin Neoplasms/metabolism , Tumor Cells, Cultured
10.
Int J Hyperthermia ; 16(2): 171-81, 2000.
Article in English | MEDLINE | ID: mdl-10763745

ABSTRACT

The objectives of this study were to quantity and compare the activities of a minimal heat shock (HS) promoter and other promoters used in gene therapy applications, and to identify strategies to amplify the heat inducibility of therapeutic genes. Human tumour cells were transiently or stably transfected with the HS promoter driving expression of reporter genes. HS promoter activity was induced transiently, with maximum activity 16-24 h after HS, and was dependent on temperature. The activity of the minimal HS promoter was similar, after 42 degrees C HS for 1 h, to that of the cytomegalovirus (CMV) promoter. To determine if the HS promoter could be used to activate a second conditional promoter, cells were transiently transfected with vectors containing both the HS and human immunodeficiency virus type 1 (HIV1) promoters. When the IL-2 gene was placed downstream of the HIV1 promoter. IL-2 production was temperature-independent. The addition of the HIV tat gene downstream of the HS promoter caused IL-2 to be induced more than 3 fold after a single 42 degrees C HS. These data indicate that the minimal HS promoter, following activation by clinically attainable temperatures (< or = 42 degrees C), can drive expression of therapeutic genes at levels comparable to the CMV promoter and be used in conjunction with a second conditional promoter to drive temperature-dependent, gene expression.


Subject(s)
Genetic Therapy , Genetic Vectors , Hyperthermia, Induced , Genes, tat , Green Fluorescent Proteins , HIV-1/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Interleukin-2/genetics , Luminescent Proteins/genetics , Promoter Regions, Genetic , Tumor Cells, Cultured
11.
Cancer Gene Ther ; 6(5): 437-46, 1999.
Article in English | MEDLINE | ID: mdl-10505854

ABSTRACT

Replication-deficient adenovirus vectors are efficient vehicles for delivering therapeutic genes into mammalian cells. However, the high doses required to produce effective gene transfer in vivo can also cause unwanted cellular toxicity. To improve replication-deficient adenovirus transgene expression while minimizing adverse reactions, we have tested polycationic compounds for their ability to enhance adenovirus adsorption. We demonstrate increased transgene expression after mixing adenovirus preparations with polycations, cationic lipids, and CaCl2 prior to transduction in vitro. An E1-deleted adenovirus vector was admixed with various polycations, and beta-galactosidase (beta-gal) activity was evaluated. The optimal polycation concentrations for augmenting adenovirus-mediated gene transfer were 5-10 microg/mL polybrene, 400 microg/mL protamine sulfate, 10 microg/mL N-(1-[2,3-dioleoyloxy]propyl)-N,N,N-trimethylammonium methylsulfate (DOTAP), 2.5 microg/mL Lipofectamine, and 62.5 mM CaCl2. Polycations enhanced beta-gal expression in three of six established cell lines. Similar results were obtained using primary tumor cell cultures, where beta-gal expression was increased 1.5- to 10.7-fold (mean = 3.6) by polybrene, 1.8- to 7.5-fold (mean = 3.4) by DOTAP, and 2.3- to 10.4-fold (mean = 4.8) by protamine sulfate. Adenovirus transduction efficiency in two primary leukemia isolates was improved by 3- and 4.5-fold. We were unable to demonstrate any benefit when adenovirus was admixed with protamine sulfate prior to intratumoral injection in a xenogeneic severe combined immunodeficient mouse melanoma tumor model. Further studies will determine whether polycations can improve intratumoral gene transfer.


Subject(s)
Adenoviridae/genetics , Lipids/pharmacology , Transduction, Genetic , Transgenes , Animals , Cations , Defective Viruses/genetics , Genetic Therapy , Humans , Mice , Neoplasms/therapy , Protamines/pharmacology , Tumor Cells, Cultured
12.
J Clin Oncol ; 17(10): 3313-23, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506635

ABSTRACT

PURPOSE: We have completed a phase I study, followed by three phase I/II studies, in patients with metastatic melanoma, renal cell carcinoma (RCC), and sarcoma in order to evaluate the safety, toxicity, and antitumor activity of Leuvectin (Vical Inc, San Diego, CA), a gene transfer product containing a plasmid encoding human interleukin (IL)-2 formulated with the cationic lipid 1, 2-dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide/dioleyl-phosphatidyl-ethanolamine (DMRIE/DOPE) and administered intratumorally. PATIENTS AND METHODS: Twenty-four patients were treated in the phase I study. Leuvectin doses were 10 microg, 30 microg, or 300 microg weekly for 6 weeks. In three subsequent phase I/II studies, a total of 52 patients (18 with melanoma, 17 with RCC, and 17 with sarcoma) were treated with further escalating doses of Leuvectin: 300 microg twice a week for 3 weeks, 750 microg weekly for 6 weeks, and 1,500 microg weekly for 6 weeks. RESULTS: There were no drug-related grade 4 toxicities and only one grade 3 toxicity, but the majority of patients experienced mild constitutional symptoms after treatment. In the phase I/II studies, 45 patients were assessable for response (14 with RCC, 16 with melanoma, and 15 with sarcoma). Two patients with RCC and one with melanoma have achieved partial responses lasting from 16 to 19 months and continuing. In addition, two RCC, three melanoma, and six sarcoma patients had stable disease lasting from 3 to 18 months and continuing. The plasmid was detected by polymerase chain reaction assay in the posttreatment samples of 29 of 46 evaluated patients. Immunohistochemistry studies on serial biopsy specimens showed increased IL-2 expression and CD8(+) infiltration after treatment in the tumor samples of several patients (12 and 16, respectively). CONCLUSION: Direct intratumoral injection of Leuvectin is a safe and possibly effective immunotherapeutic approach in the treatment of certain tumor types.


Subject(s)
Carcinoma, Renal Cell/therapy , Gene Transfer Techniques , Genetic Therapy , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Melanoma/therapy , Sarcoma/therapy , Skin Neoplasms/therapy , Adult , Aged , CD8 Antigens/analysis , Carcinoma, Renal Cell/pathology , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Interleukin-2/genetics , Interleukin-2/pharmacokinetics , Kidney Neoplasms/pathology , Lipids/genetics , Lipids/therapeutic use , Male , Melanoma/pathology , Middle Aged , Plasmids/genetics , Polymerase Chain Reaction , Quaternary Ammonium Compounds/therapeutic use , Sarcoma/pathology , Skin Neoplasms/pathology
13.
Cancer Chemother Pharmacol ; 44(4): 303-6, 1999.
Article in English | MEDLINE | ID: mdl-10447577

ABSTRACT

PURPOSE: Chemotherapeutic regimens that utilize fluorouracil, cytarabine, and doxorubicin have been shown to cause a dermatologic syndrome known as hand-foot syndrome, or palmar-plantar erythrodysesthesia syndrome (PPES). Pegylated liposomal doxorubicin has proven effective in the treatment of AIDS-related Kaposi's sarcoma, ovarian cancer refractory to platinum and paclitaxel therapies, and metastatic breast cancer. In a study of the treatment of refractory epithelial cell ovarian cancers with lipozomal doxorubicin utilizing intravenous doses of 50 mg/m(2) every 3 weeks, grade 3 PPES was observed in 29% of patients (10/35) and required dose reductions and/or dose delay after a median of three therapy cycles. METHODS: Current methods to prevent pegylated liposomal doxorubicin-induced PPES include dose reduction, lengthening of the drug administration interval and ultimately, drug withdrawal. Topical 99% dimethylsulfoxide (DMSO) also has shown strong activity in treating tissue extravasation reactions during intravenous administration of doxorubicin. RESULTS: Two patients undergoing chemotherapy with pegylated liposomal doxorubicin, 50 mg/m(2) every 4 weeks, developed grade 3 PPE after three cycles. Their PPES resolved over a period of 1 to 3 weeks while receiving topical 99% DMSO four times daily for 14 days. CONCLUSIONS: While these results are promising, patients must be treated in a prospective study of this topical DMSO formulation to definitively document its therapeutic efficacy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/adverse effects , Dimethyl Sulfoxide/therapeutic use , Doxorubicin/adverse effects , Erythema/chemically induced , Erythema/drug therapy , Foot , Hand , Paresthesia/chemically induced , Paresthesia/drug therapy , Administration, Topical , Female , Humans , Liposomes , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Syndrome
14.
J Cancer Res Clin Oncol ; 125(5): 292-6, 1999.
Article in English | MEDLINE | ID: mdl-10359134

ABSTRACT

The therapeutic benefit of adding interferon alpha (IFNalpha) to established single-agent and combination chemotherapy regimens for the treatment of metastatic melanoma has not been proven. We designed the present study to estimate the response rate of IFNalpha, dacarbazine, cisplatin and tamoxifen in patients who had not been treated with systemic therapy for advanced disease. Using a schedule similar to that which had previously been shown to favor IFNalpha plus dacarbazine over dacarbazine alone, we treated patients with an "induction" regimen of IFNalpha, 15 mU m(-2) day(-1) intravenously 5 days/week for 3 weeks. Following induction, schedules of IFNalpha, 5 mU m(-2) day(-1) subcutaneously three times a week, and tamoxifen, 10 mg orally twice a day, were begun. Dacarbazine, 250 mg m(-2) day(-1) and cisplatin 33 mg m(-2) day(-1) for 3 consecutive days were repeated every 4 weeks, and subcutaneous IFNalpha and oral tamoxifen were continued until the discontinuation of chemotherapy. We treated 25 patients (18 men and 7 women, median age 52 years) and observed only 1 objective response (response rate 4%, 95% confidence interval 0.1%-20%). The toxicities of the regimen consisted of moderate myelosuppression and constitutional side-effects. On the basis of the low antitumor activity of this regimen, we do not recommend it for further study or for use as standard therapy of metastatic melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Melanoma/secondary , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Drug Administration Schedule , Female , Humans , Interferon-alpha/administration & dosage , Male , Middle Aged , Southwestern United States , Tamoxifen/administration & dosage , Treatment Outcome
15.
J Med Chem ; 42(3): 510-4, 1999 Feb 11.
Article in English | MEDLINE | ID: mdl-9986721

ABSTRACT

A set of 20 2-cyanoaziridine-1-carboxamides was synthesized from 2-cyanoaziridine and appropriate isocyanates. These compounds were active against a variety of solid and hematological tumor cells in culture, including strains resistant to doxorubicin and mitoxantrone. Their potencies in these assays correlated with the lipophilicity of substituents. The N-phenyl derivative was more potent and equally effective to imexon, a cyclized 2-cyanoaziridine-1-carboxamide of clinical interest, against cloned fresh human tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Antineoplastic Agents/chemistry , Aziridines/chemistry , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Curr Opin Mol Ther ; 1(2): 158-76, 1999 Apr.
Article in English | MEDLINE | ID: mdl-11715940

ABSTRACT

Infectious disease, heart disease, cancer, autoimmunity, genetic defects and even traumatic injury may someday be treated with gene therapy and gene transfer strategies. The potential impact of this new technology on human disease has produced optimism and expectation for scientists and lay people alike. As more effort is directed at harvesting the potential of this technology it has become clear that the success or failure of gene therapy will hinge on our ability to manipulate and control the process of gene transfer in somatic cells. Today, somatic gene transfer is accomplished using either viral or non-viral gene transfer methods. The benefits and limitations of each system are aggressively being investigated to determine which characteristics are most compatible with safe and reliable gene transfer. Gene transfer with cationic lipid/plasmid DNA complexes (cationic lipoplexes) has become a popular means of delivering therapeutic genes and is being tested in preclinical and clinical trials. Cationic lipoplexes are easy and inexpensive to produce, they are composed of non-toxic and non-immunogenic precursor, and they have the potential of delivering large polynucleotides into somatic cells. Additionally, these reagents are easily manipulated in the laboratory to incorporate novel biological functions or to produce new formulations that can be screened for in vivo gene transfer activity. The last few years has seen many advances in our understanding of molecular and biological factors that influence cationic lipid-mediated gene transfer. In this review we discuss recent developments in the field of cationic lipid-mediated gene transfer with emphasis on in vivo application where possible. We will consider new discoveries concerning the molecular and cellular events that control the uptake, transit and expression of lipoplexes in somatic cells. Recent biodistribution and pharmacokinetic studies and current concepts regarding the toxicity and immunogenicity of cationic lipoplexes will also be discussed. We also survey some of the many preclinical and clinical trials using cationic lipid-mediated gene transfer, with emphasis on cancer applications.


Subject(s)
Lipids/administration & dosage , Transfection , Animals , Cations , Humans
17.
Cancer Gene Ther ; 5(2): 119-26, 1998.
Article in English | MEDLINE | ID: mdl-9570303

ABSTRACT

Cancer vaccines that utilize genetically modified tumor cells require gene transfer methods capable of producing immunostimulatory doses of transgenes from fresh or short-term cultures of human tumor cells. Our studies optimize in vitro transfection of primary tumor cells using cationic lipids and a plasmid encoding the gene for human interleukin-2 (IL-2). Established tumor cell lines produced 10- to 100-fold more IL-2 than did fresh or short-term tumor cultures as measured by enzyme-linked immunoabsorbent analysis. Importantly, transfection of primary tumor cells produced immunostimulatory levels of IL-2 as determined by increased thymidine incorporation by autologous peripheral blood mononuclear cells and lymphokine-activated killer cell activity. IL-2 secretion by tumor cells persisted for at least 30 days post-transfection and was unaffected by freeze thawing or irradiation to 8000 rads. Multiple solid tumor types were successfully transfected, but normal blood mononuclear cells and leukemic blasts were resistant to transfection. Enzyme-linked immunoabsorbent analysis of the amount of IL-2 secreted into the medium by transfected tumor cells correlated with the percentage of tumor cells expressing intracellular IL-2 as measured by flow cytometry. Plasmids utilizing a cytomegalovirus promoter yielded superior transfection efficiencies compared with plasmids containing a Rous sarcoma virus promoter. These results suggest that a clinical vaccine trial using autologous tumor cells genetically modified to secrete IL-2 is feasible in patients with solid tumors.


Subject(s)
Cancer Vaccines/genetics , DNA/administration & dosage , Lipids/administration & dosage , Neoplasms/immunology , Plasmids , Avian Sarcoma Viruses/genetics , Cytomegalovirus/genetics , Escherichia coli/genetics , Humans , Interleukin-2/biosynthesis , Killer Cells, Lymphokine-Activated/immunology , Neoplasms/pathology , Neoplasms/therapy , Promoter Regions, Genetic , Transfection , Tumor Cells, Cultured , beta-Galactosidase/genetics
19.
Cancer Chemother Pharmacol ; 40(3): 209-14, 1997.
Article in English | MEDLINE | ID: mdl-9219503

ABSTRACT

PURPOSE: To test a number of established human tumor cell lines and early passage breast cancer (UACC2150) and melanoma cells (UACC1273) for growth in the scid mouse and the tumors' response to conventional chemotherapeutic drugs. METHODS: Established melanoma (A375, C81-61), colon (SW480), lung (A549), lymphomoblastoid leukemia (LCL-B), promyelocytic leukemia (HL60), prostate (PC-3, DU145), and breast (MCF7) cell lines were injected at subcutaneous (s.c.), intraperitoneal (i.p.), or mammary fat pad (MFP) sites. Tumor volume growth curves and survival curves were established for the various tumor cell lines. Carmustine (BCNU), cisplatin (CDDP), cyclophosphamide (CPA), doxorubicin, dacarbazine (DTIC), tamoxifen and vincristine were injected s.c. or i.p.. The chemotherapeutic drug effects on tumor volumes and survival were determined. RESULTS: Tumor growth occurred with each cell type. After i.p. injection, 90% mortality occurred within 26 to 60 days except for the early passage melanoma cell line UACC1273 with which mortality occurred within approximately 90 days. In the MCF7 breast model, treatment with tamoxifen (P < 0.001) and CPA (P < 0.0001) resulted in significant tumor growth delay compared with control groups. BCNU and CDDP resulted in significant tumor growth delays relative to control in SW480 colon cancer (P < 0.0014) and A375 melanoma (P < 0.0001) models, respectively. CPA and doxorubicin improved survival in the HL60 leukemia model (P = 0.0018). CONCLUSIONS: These scid mouse human tumor models appear to reflect the clinical situation in that clinically active chemotherapeutic drugs are similarly active in the scid mouse models. Therefore, the scid mouse models may be useful for testing new chemotherapeutic agents against various human cancer types.


Subject(s)
Antineoplastic Agents/pharmacology , Disease Models, Animal , Neoplasms, Experimental/drug therapy , Severe Combined Immunodeficiency/drug therapy , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Drug Administration Schedule , Female , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Severe Combined Immunodeficiency/mortality , Severe Combined Immunodeficiency/pathology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
20.
Cancer Gene Ther ; 4(1): 42-50, 1997.
Article in English | MEDLINE | ID: mdl-9012450

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a potent immunosuppressive cytokine produced by many tumor cells. Secretion of TGF-beta by malignant cells may therefore be a mechanism by which tumor cells escape destruction by tumor-specific T lymphocytes. In order to evaluate the role of tumor-derived TGF-beta on tumor progression, we have inhibited the production of this cytokine by introducing a gene encoding antisense TGF-beta1 into the EMT6 murine mammary tumor cell line using a retroviral vector (Las-TGF-beta1SN). EMT6 cells transduced with this vector (EMT6as-TGF-beta1) stably expressed the antisense gene and secreted 52% less TGF-beta than did tumor cells transduced with the backbone vector alone. Supernatant fluid recovered from tumor cells expressing the antisense TGF-beta1 gene also exhibited a decreased capacity to inhibit alloantigen-specific cytotoxic T-cell responses in vitro. Furthermore, tumor growth in mice injected with EMT6as-TGF-beta1 tumor cells was inhibited compared to mice injected with control tumor cells. These results demonstrate that expression of antisense TGF-beta1 by transduced EMT6 cells decreases their tumorigenicity and suggest that this approach of eliminating immune suppression is a potentially useful strategy to enhance antitumor responses.


Subject(s)
Gene Expression , Mammary Neoplasms, Experimental/pathology , Oligonucleotides, Antisense/genetics , Transforming Growth Factor beta/genetics , Transgenes , Animals , Cell Cycle/genetics , Cell Cycle/immunology , Cell Division/genetics , Cell Division/immunology , Female , Immune Tolerance , Mice , Mice, Inbred BALB C , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...