Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Med ; 27(10): 1789-1796, 2021 10.
Article in English | MEDLINE | ID: mdl-34608333

ABSTRACT

Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-based intralesional oncolytic immunotherapy approved for the treatment of unresectable melanoma. The present, ongoing study aimed to estimate the treatment effect of neoadjuvant T-VEC on recurrence-free survival (RFS) of patients with advanced resectable melanoma. An open-label, phase 2 trial (NCT02211131) was conducted in 150 patients with resectable stage IIIB-IVM1a melanoma who were randomized to receive T-VEC followed by surgery (arm 1, n = 76) or surgery alone (arm 2, n = 74). The primary endpoint was a 2-year RFS in the intention-to-treat population. Secondary and exploratory endpoints included overall survival (OS), pathological complete response (pCR), safety and biomarker analyses. The 2-year RFS was 29.5% in arm 1 and 16.5% in arm 2 (overall hazard ratio (HR) = 0.75, 80% confidence interval (CI) = 0.58-0.96). The 2-year OS was 88.9% for arm 1 and 77.4% for arm 2 (overall HR = 0.49, 80% CI = 0.30-0.79). The RFS and OS differences between arms persisted at 3 years. In arm 1, 17.1% achieved a pCR. Increased CD8+ density correlated with clinical outcomes in an exploratory analysis. Arm 1 adverse events were consistent with previous reports for T-VEC. The present study met its primary endpoint and estimated a 25% reduction in the risk of disease recurrence for neoadjuvant T-VEC plus surgery versus upfront surgery for patients with resectable stage IIIB-IVM1a melanoma.


Subject(s)
Biological Products/administration & dosage , Immunotherapy , Melanoma/therapy , Neoadjuvant Therapy , Adult , Aged , Biological Products/immunology , Combined Modality Therapy , Disease-Free Survival , Female , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/virology , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Neoplasm Recurrence, Local/virology , Neoplasm Staging , Oncolytic Virotherapy/trends , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology
3.
J Immunother Cancer ; 4: 60, 2016.
Article in English | MEDLINE | ID: mdl-27777770

ABSTRACT

BACKGROUND: Anti-PD-1 therapy is increasingly used in various advanced malignancies. Patients with baseline organ dysfunction are largely excluded from clinical trials. Therefore it is unclear whether anti-PD-1 therapy is safe or effective in this setting. Further, these patients are often not candidates for other anti-cancer therapies, highlighting their need for active treatment options. METHODS: We performed a retrospective analysis of patients from multiple centers with advanced solid tumors and baseline organ dysfunction who received anti-PD-1 therapy. Organ dysfunction was defined as cardiac (left ventricular ejection fraction ≤45 %), renal (creatinine ≥2 mg/dL or GFR ≤30 ml/min) or hepatic dysfunction (evidence of cirrhosis on imaging or AST, ALT or bilirubin ≥3x ULN). We assessed change in organ dysfunction, immune related adverse events (irAEs), response rate, progression free survival (PFS) and overall survival (OS). RESULTS: We identified 27 patients eligible for inclusion with the following diseases: renal cell carcinoma (n = 8), melanoma (10), non-small cell lung cancer (3), small cell lung cancer (2) and urothelial bladder cancer (4). Baseline organ dysfunction included renal dysfunction (n = 17), hepatic dysfunction (7), cardiac dysfunction (11), including >1 organ dysfunction (8). Worsening organ dysfunction requiring hospitalization or dose delays occurred in 8 patients (30 %) although in most cases this was thought not-drug related and resolved with supportive care. Grade 3 irAEs occurred in 2 pts (7 %; hepatitis and colitis). Thirteen of 27 patients had ongoing treatment benefit (objective response or stable disease) at data collection (48 %). Eleven patients had primary progressive disease (41 %), 11 had stable disease (41 %), 4 had partial responses (15 %), and one had a complete response (4 %). Overall, median PFS was 168 days. Median OS was not reached. CONCLUSIONS: In our experience, anti-PD-1 agents in this group of patients with cardiac, hepatic or renal dysfunction were associated with tolerable irAEs and infrequent manageable worsening of organ dysfunction. Further, objective responses and prolonged PFS were observed in a number of patients. Thus, patients with baseline organ dysfunction may be considered for anti-PD-1 therapy with appropriate clinical monitoring.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Heart/drug effects , Kidney/drug effects , Liver/drug effects , Molecular Targeted Therapy/adverse effects , Programmed Cell Death 1 Receptor/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Cardiotoxicity , Female , Heart/physiopathology , Heart Function Tests , Humans , Kidney/physiopathology , Kidney Function Tests , Liver/physiopathology , Liver Function Tests , Male , Middle Aged , Neoplasm Staging , Neoplasms/complications , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/mortality , Positron Emission Tomography Computed Tomography , Retrospective Studies , Treatment Outcome
4.
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
5.
Ann Oncol ; 26(7): 1481-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25899782

ABSTRACT

BACKGROUND: The use of Escherichia coli purine nucleoside phosphorylase (PNP) to activate fludarabine has demonstrated safety and antitumor activity during preclinical analysis and has been approved for clinical investigation. PATIENTS AND METHODS: A first-in-human phase I clinical trial (NCT 01310179; IND 14271) was initiated to evaluate safety and efficacy of an intratumoral injection of adenoviral vector expressing E. coli PNP in combination with intravenous fludarabine for the treatment of solid tumors. The study was designed with escalating doses of fludarabine in the first three cohorts (15, 45, and 75 mg/m(2)) and escalating virus in the fourth (10(11)-10(12) viral particles, VP). RESULTS: All 12 study subjects completed therapy without dose-limiting toxicity. Tumor size change from baseline to final measurement demonstrated a dose-dependent response, with 5 of 6 patients in cohorts 3 and 4 achieving significant tumor regression compared with 0 responsive subjects in cohorts 1 and 2. The overall adverse event rate was not dose-dependent. Most common adverse events included pain at the viral injection site (92%), drainage/itching/burning (50%), fatigue (50%), and fever/chills/influenza-like symptoms (42%). Analysis of serum confirmed the lack of systemic exposure to fluoroadenine. Antibody response to adenovirus was detected in two patients, suggesting that neutralizing immune response is not a barrier to efficacy. CONCLUSIONS: This first-in-human clinical trial found that localized generation of fluoroadenine within tumor tissues using E. coli PNP and fludarabine is safe and effective. The pronounced effect on tumor volume after a single treatment cycle suggests that phase II studies are warranted. CLINICALTRIALSGOV IDENTIFIER: NCT01310179.


Subject(s)
Escherichia coli/enzymology , Genetic Therapy , Genetic Vectors/therapeutic use , Neoplasms/genetics , Neoplasms/therapy , Purine-Nucleoside Phosphorylase/administration & dosage , Vidarabine/analogs & derivatives , Adenoviridae/genetics , Aged , Aged, 80 and over , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Injections, Intralesional , Male , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Prognosis , Purine-Nucleoside Phosphorylase/genetics , Tumor Cells, Cultured , Vidarabine/therapeutic use
6.
J Am Acad Dermatol ; 67(6): 1375-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940405

ABSTRACT

Nonmalignant cutaneous findings associated with the use of vemurafenib have only recently been described in the literature. Patients receiving vemurafenib have exhibited cutaneous reactions including prominent follicular plugging, hand-foot skin reaction, exuberant seborrheic dermatitis-like hyperkeratosis of the face, keratosis pilaris, and diffuse spiny follicular hyperkeratosis. Many of these nonmalignant cutaneous findings are associated with abnormal follicular keratinization thought to be secondary to abnormal signaling of the mitogen-activated protein kinase pathway that occurs with the use of BRAF inhibitors. Whether underlying Ras mutations affect this abnormal signaling as in malignant lesions is still unknown. Different therapeutic options exist for these patients that may result in significant improvement in some of these nonmalignant cutaneous findings. Conservative treatment should focus on topical therapies such as topical retinoids or topical steroids. However, systemic therapies such as concomitant oral retinoids or MEK inhibitors should be considered for more severe or refractory cutaneous findings.


Subject(s)
Drug Eruptions/etiology , Indoles/adverse effects , Melanoma/drug therapy , Melanoma/secondary , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Sulfonamides/adverse effects , Drug Eruptions/therapy , Humans , Indoles/therapeutic use , Sulfonamides/therapeutic use , Vemurafenib
7.
Ann Oncol ; 22(4): 787-793, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20855467

ABSTRACT

BACKGROUND: Docosahexaenoic acid-paclitaxel (DHA-paclitaxel, Taxoprexin(®)) is made by covalently conjugating the essential fatty acid DHA to the paclitaxel molecule. Preclinical studies of DHA-paclitaxel have demonstrated increased activity relative to paclitaxel and the potential for an improved therapeutic ratio. In the present study, the efficacy and toxicity profiles of DHA-paclitaxel were compared with those of dacarbazine. METHODS: In this study, 393 chemonaive patients with metastatic melanoma were randomly assigned to receive either DHA-paclitaxel at a starting dose of 900 mg/m(2) IV on day 1 every 3 weeks or dacarbazine at a starting dose of 1000 mg/m(2) IV on day 1 every 3 weeks. The primary end point of the study was the comparison of overall survival (OS). RESULTS: No significant difference in OS was noted between patients in the DHA-paclitaxel and dacarbazine arms. Similarly, there were no significant differences in response rate, duration of response, time to progression, and time to treatment failure between the two drugs. Safety results of the two drugs were as predicted from prior studies. Myelosuppression was more common with DHA-paclitaxel. CONCLUSIONS: DHA-paclitaxel was not superior to dacarbazine. We conclude that further studies with the drug on an every 3-week schedule in melanoma are not warranted.


Subject(s)
Dacarbazine/therapeutic use , Melanoma/drug therapy , Paclitaxel/analogs & derivatives , Adolescent , Adult , Aged , Aged, 80 and over , Dacarbazine/adverse effects , Disease Progression , Disease-Free Survival , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Paclitaxel/adverse effects , Paclitaxel/therapeutic use
8.
J Biol Inorg Chem ; 6(4): 359-66, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11372195

ABSTRACT

The reaction of Mo(O)2(acac)2, H2L (2,2'-dimercaptobiphenyl), and NEt3 produced the mononuclear Mo(V) complex Et3NH[Mo(O)(L)2] (1). Molybdenum mono-oxo tetrathiolate complexes such as 1 are studied as potential structural or functional models for pyranopterin-containing molybdoenzymes. Complex 1 has been crystallographically characterized. The solid-state structure reveals that the molybdenum ion sits within a cleft formed by the biphenyl backbone of the ligands, providing some steric protection. In addition, there is a hydrogen bond between the amine hydrogen of [Et3NH]+ and one of the thiolate sulfur atoms. A difference in solution reactivity between 1 and a derivative without a hydrogen-bonding counterion suggests that hydrogen bonding occurs in solution also. There are two short S-S contacts and small S-Mo-S angles in the structure of 1 that may reflect a slight bonding interaction. Such short S-S distances and small angles have been found in a couple of other Mo-thiolate complexes and in many of the molybdoenzyme crystal structures. Further characterization of 1 by EPR, IR, and UV-vis spectroscopies, as well as by cyclic voltammetry, is discussed and compared to known Mo(V)-oxo-tetrathiolate complexes as well as to relevant molybdoenzyme data. Reactions to generate Mo(VI) complexes from 1 resulted in net oxidation at the ligand to form its disulfide derivative, which dissociated from the metal center. This result suggests that modifications to the ligand to prevent this process are needed.


Subject(s)
Molybdenum/chemistry , Organometallic Compounds/chemistry , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Hydrogen Bonding , Ligands , Models, Molecular
9.
J Immunother ; 23(3): 379-86, 2000.
Article in English | MEDLINE | ID: mdl-10838667

ABSTRACT

MART-1, a melanoma antigen recognized by T cells-1, is a melanocyte lineage-differentiation antigen expressed only in melanocytes and melanoma cells. This protein is recognized by many T-lymphocyte lines that are human leukocyte antigen (HLA)-A2 restricted and melanoma reactive. These observations have culminated in an array of clinical trials of MART-1 immunization using recombinant viruses or MART-1 immunodominant peptides. Polynucleotide immunization is a promising alternative to recombinant viral vaccines that allows delivery of the full-length cDNA encoding all potential peptide epitopes in a vector that is uncompromised by anti-viral immunity. In preparation for a phase I clinical trial of MART-1 polynucleotide immunization in patients with resected melanoma who were at significant risk for recurrence, the authors constructed a plasmid DNA encoding the MART-1 cDNA under transcriptional regulatory control of the cytomegalovirus immediate early promoter-enhancer and partially deleted intron A. This plasmid directs high-level MART-1 expression in transduced myoblasts and maturing myocytes diffusely throughout the cytoplasm. Immunization of mice with this construct by intramuscular injection elicited MART-1-specific immune responses in all animals. Previous trials of MART-1 immunization have been unable to examine the humoral immune response to MART-1 because of a lack of sufficient, highly purified protein. We have produced and purified Escherichia coli recombinant MART-1 protein using a glutathione-S-transferase fusion protein expression system. Protein staining of a sodium dodecyl sulfate-polyacrylamide gel electrophoresis revealed a band of MART-1 protein at approximately 20 kD; and Western immunoblotting with an anti-MART-1 monoclonal antibody confirmed a doublet at approximately 20 kD. These findings are consistent with previous reports using different expression systems for recombinant MART-1. This protein preparation functioned well in enzyme-linked immunosorbent assays (ELISAs) to detect anti-MART-1 antibody responses in a mouse model; and a panel of healthy donor human sera showed minimal binding to ELISA plates coated with the protein, supporting its utility in monitoring human anti-MART-1 antibody responses. The glutathione-S-transferase fusion method yielded approximately 200 micrograms MART-1 per 2-L bacterial culture, enough to coat 100 ELISA plates.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/isolation & purification , Cancer Vaccines/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , Vaccines, DNA/immunology , Animals , Antibody Formation , Cell Line , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Female , Immunohistochemistry , MART-1 Antigen , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Plasmids , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
10.
Pediatrics ; 105(5): 1096-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10790468

ABSTRACT

PURPOSE: To determine the structural and functional integrity of the brain in a sample of nonretarded individuals diagnosed with fetal alcohol syndrome. SUBJECTS: The sample consisted of nineteen individuals who met the diagnostic criteria for fetal alcohol syndrome. METHODS: Intellectual function was assessed using the Wechsler Adult Intelligence Scale-Revised. Structural integrity of the brain was assessed using magnetic resonance imaging whereas functional integrity was assessed using positron emission tomography and (18)F-fluorodeoxyglucose. RESULTS: The mean Full Scale IQ was 80. 2 (range: 66-92). Only 1 magnetic resonance imaging was found to be abnormal. This abnormality was found for the subject with the lowest IQ. Decreases in relative regional cerebral metabolic rates were found in 5 brain regions comprising the thalamus and basal ganglia. CONCLUSION: These results when coupled with previous findings suggest a continuum of neuropathology in fetal alcohol syndrome. For cases with relatively mild intellectual deficits, the cause of the deficit is at a micro-level rather than a macro-level.


Subject(s)
Fetal Alcohol Spectrum Disorders/pathology , Fetal Alcohol Spectrum Disorders/physiopathology , Adolescent , Adult , Brain/pathology , Brain/physiopathology , Female , Humans , Magnetic Resonance Imaging , Male , Tomography, Emission-Computed
11.
J Gene Med ; 2(2): 135-40, 2000.
Article in English | MEDLINE | ID: mdl-10809147

ABSTRACT

BACKGROUND: We have previously reported that intramuscular, intradermal or epidermal gene gun administration of a plasmid encoding carcinoembryonic antigen (CEA) under transcriptional regulatory control of the cytomegalovirus (CMV) early promoter/enhancer elicits CEA-specific humoral and cellular immune responses in mice with resultant immunoprotection against challenge with syngeneic, CEA-expressing colon adenocarcinoma cells. METHODS: In the present work, we examine the ability of this DNA vaccine construct (pCEA) to elicit CEA-specific immunity following intrasplenic administration. Groups of mice were immunized with pCEA by intrasplenic or intramuscular injection. Six weeks later, mice were evaluated for the presence of anti-CEA humoral responses and were challenged with syngeneic, CEA-expressing colon carcinoma cells. RESULTS: Intrasplenic administration of pCEA produced a frequency of CEA-specific antibody responses comparable to that elicited by intramuscular pCEA inoculation. Both intrasplenic and intramuscular administration of pCEA generated IgG2a antibody responses to CEA, consistent with the induction of T helper-1-biased immune responses. In addition, partial immunoprotection against tumor challenge was observed after a single plasmid DNA dose by either route of administration. Subsequent studies revealed that antibody responses to intrasplenic DNA vaccination are dose and schedule dependent. CONCLUSION: These findings support future investigations of DNA vaccination strategies that specifically promote the uptake of plasmid by splenocytes.


Subject(s)
Carcinoembryonic Antigen/immunology , Colonic Neoplasms/prevention & control , Vaccines, DNA/pharmacology , Animals , Antibody Formation , Carcinoembryonic Antigen/genetics , Colonic Neoplasms/immunology , Cytomegalovirus/genetics , Female , Gene Transfer Techniques , Genetic Vectors , Injections , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Spleen , Tumor Cells, Cultured , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
12.
Cancer Gene Ther ; 7(3): 446-55, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10766351

ABSTRACT

Evaluation of immunotherapy strategies in mouse models of carcinoma is hampered by the limited number of known murine tumor antigens (Ags). Although tumor Ags can be identified based on cytotoxic T-cell activation, this approach is not readily accomplished for many tumor types. We applied an alternative strategy based on a humoral immune response, SEREX, to the identification of tumor Ags in the murine colon adenocarcinoma cell line MC38. Immunization of syngeneic C57BL/6 mice with MC38 cells by three different methods induced a protective immune response with concomitant production of anti-MC38 antibodies. Immunoscreening of an MC38-derived expression library resulted in the identification of the endogenous ecotropic leukemia virus envelope (env) protein and the murine ATRX protein as candidate tumor Ags. Northern blot analysis demonstrated high levels of expression of the env transcript in MC38 cells and in several other murine tumor cell lines, whereas expression in normal colonic epithelium was absent. ATRX was found to be variably expressed in tumor cell lines and in normal tissue. Further analysis of the expressed env sequence indicated that it represents a nonmutated tumor Ag. Polynucleotide immunization with DNA encoding the env polypeptide resulted in strong and specific antibody responses to this self Ag in all immunized mice. Thus, SEREX offers a rapid means of identifying tumor Ags in murine cancer models.


Subject(s)
Adenocarcinoma/immunology , Antigens, Neoplasm/isolation & purification , DNA Helicases , Nuclear Proteins , Adenocarcinoma/chemistry , Animals , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Base Sequence , Blotting, Southern , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Products, env/biosynthesis , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, env/isolation & purification , Hemolytic Plaque Technique , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Neoplasm Transplantation , Polynucleotides/administration & dosage , Polynucleotides/immunology , RNA, Messenger/biosynthesis , Sequence Analysis, DNA , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Cells, Cultured/transplantation , X-linked Nuclear Protein
13.
Clin Cancer Res ; 6(1): 34-41, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10656429

ABSTRACT

Carcinoembryonic antigen (CEA) is a well-characterized oncofetal glycoprotein whose overexpression by human adenocarcinomas has been a target for cancer immunotherapy. Limited information is available regarding the ability of patients to mount an antibody response to this self-antigen following vaccination. Recombinant vaccinia viruses encoding full-length or internally deleted cDNAs for human CEA were used to vaccinate 32 patients with CEA-expressing adenocarcinomas, predominantly of colorectal origin. CEA-specific autoantibodies were induced by vaccination in 7 of 32 patients. None of the patients had CEA antibodies detected before vaccination. CEA specificity of the antibodies initially identified by ELISA was confirmed by competitive inhibition analysis as well as recognition of recombinant CEA produced in baculovirus-infected insect cell cultures and human cell cultures by Western blot. The CEA autoantibodies were predominantly IgG1, with a minority of patients also demonstrating IgM autoantibodies. CEA antibodies were of low titer and low avidity, based on competitive inhibition assays. These autoantibodies did not affect clinical serum CEA protein quantitation. Furthermore, elevated serum CEA levels commonly encountered in patients with advanced adenocarcinoma did not hinder detection of low avidity polyclonal CEA antibodies. CEA antibodies such as those induced in these pilot trials are projected to have modest antitumor activity. Thus, additional Phase I/II trials of recombinant vaccinia-CEA with alternative prime-boost approaches and/or augmentation strategies are warranted in an effort to enhance the frequency and avidity of CEA-specific autoantibodies and cytolytic T cells before Phase III trials.


Subject(s)
Adenocarcinoma/immunology , Autoantibodies/blood , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/immunology , Immunoglobulin G/blood , Vaccines, Synthetic/immunology , Adenocarcinoma/therapy , Antibodies, Monoclonal , Antibody Formation , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/classification , Time Factors , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/therapeutic use , Vaccinia virus
14.
Inorg Chem ; 39(25): 5697-706, 2000 Dec 11.
Article in English | MEDLINE | ID: mdl-11151370

ABSTRACT

Electronic absorption, magnetic circular dichroism, and resonance Raman spectroscopies have been used to determine the nature of oxomolybdenum-thiolate bonding in (PPh4)[MoO(SPh)4] (SPh = phenylthiolate) and (HNEt3)[MoO(SPh-PhS)2] (SPh-PhS = biphenyl-2,2'-dithiolate). These compounds, like all oxomolybdenum tetraarylthiolate complexes previously reported, display an intense low-energy charge-transfer feature that we have now shown to be comprised of multiple S-->Mo dxy transitions. The integrated intensity of this low-energy band in [MoO(SPh)4]- is approximately twice that of [MoO(SPh-PhS)2]-, implying a greater covalent reduction of the effective nuclear charge localized on the molybdenum ion of the former and a concomitant negative shift in the Mo(V)/Mo(IV) reduction potential brought about by the differential S-->Mo dxy charge donation. However, this is not observed experimentally; the Mo(V)/Mo(IV) reduction potential of [MoO(SPh)4]- is approximately 120 mV more positive than that of [MoO(SPh-PhS)2]- (-783 vs -900 mV). Additional electronic factors as well as structural reorganizational factors appear to play a role in these reduction potential differences. Density functional theory calculations indicate that the electronic contribution results from a greater sigma-mediated charge donation to unfilled higher energy molybdenum acceptor orbitals, and this is reflected in the increased energies of the [MoO(SPh-PhS)2]- ligand-to-metal charge-transfer transitions relative to those of [MoO(SPh)4]-. The degree of S-Mo dxy covalency is a function of the O identical to Mo-S-C dihedral angle, with increasing charge donation to Mo dxy and increasing charge-transfer intensity occurring as the dihedral angle decreases from 90 to 0 degree. These results have implications regarding the role of the coordinated cysteine residue in sulfite oxidase. Although the O identical to Mo-S-C dihedral angles are either approximately 59 or approximately 121 degrees in these oxomolybdenum tetraarylthiolate complexes, the crystal structure of the enzyme reveals an O identical to Mo-SCys-C angle of approximately 90 degrees. Thus, a significant reduction in SCys-Mo dxy covalency is anticipated in sulfite oxidase. This is postulated to preclude the direct involvement of coordinated cysteine in coupling the active site into efficient superexchange pathways for electron transfer, provided the O identical to Mo-SCys-C angle is not dynamic during the course of catalysis. Therefore, we propose that a primary role for coordinated cysteine in sulfite oxidase is to statically poise the reduced molybdenum center at more negative reduction potentials in order to thermodynamically facilitate electron transfer from Mo(IV) to the endogenous b-type heme.


Subject(s)
Cysteine/chemistry , Molybdenum/chemistry , Organometallic Compounds/chemistry , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Sulfhydryl Compounds/chemistry , Binding Sites , Circular Dichroism , Kinetics , Models, Molecular , Molecular Conformation , Oxidation-Reduction , Protein Conformation , Spectrophotometry , Spectrum Analysis, Raman
15.
Inorg Chem ; 39(8): 1674-9, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-12526553

ABSTRACT

Two new NiIIS4 complexes with the biphenyl-2,2'-dithiolate ligand (L) are reported. The dinuclear complex 1, [Ni2L3]2-, was formed in the reaction of 2-3 equiv of Na2L and [NiCl4]2- and the mononuclear complex [NiL2]2- (2) by using 4-10 equiv of Na2L. Complexes 1 and 2 have been crystallographically characterized. (Et4N)2[1].0.5S2Ph2, CH3CN: C60H71N3Ni2S7, triclinic, P1, a = 13.806(2) A, b = 14.267(2) A, c = 16.873(2) A, alpha = 69.263(10) degrees, beta = 69.267(8) degrees, gamma = 83.117(10) degrees, Z = 2, R1 = 0.0752 (wR2 = 0.2011). (Et4N)(Na.CH3CN)[2]: C34H39N2NaNiS4, triclinic, P1, a = 9.9570(10) A, b = 13.2670(10) A, c = 13.9560(10) A, alpha = 108.489(7) degrees, beta = 90.396(6) degrees, gamma = 103.570(4) degrees, Z = 2, R1 = 0.0390 (wR2 = 0.0995). Both complexes are square planar about the nickel ion in the solid state as well as in solution. Most Ni(II)-thiolate complexes are square planar except the tetrahedral mononuclear complexes with monodentate arylthiolate ligands that cannot force a square planar geometry. The ligand (L) has some flexibility to change its bite angle via the phenyl-phenyl bond and should not force a planar geometry on its complexes either. Therefore, it is interesting that 2 has adopted a square planar structure. Complex 2 readily converts to 1 in solution when not in the presence of excess L in a process that is presumably similar to that known for other mononuclear, bidentate ligated Ni(II) complexes. Both complexes, at least in the solid state, appear to have an inclination to bind another metal ion on one face of the complex (Ni2+ in 1, Na+ in 2). We hope to take advantage of this in future work to synthesize relevant model complexes for the active sites of the nickel-iron hydrogenases after suitable modifications are made to L.


Subject(s)
Biphenyl Compounds/chemistry , Nickel/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/chemical synthesis , Sulfur Compounds/chemistry , Sulfur Compounds/chemical synthesis , Binding Sites , Crystallography, X-Ray , Ions/chemistry , Ligands , Models, Chemical , Molecular Conformation , Molecular Structure , Sodium/chemistry
16.
Clin Cancer Res ; 5(9): 2330-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499601

ABSTRACT

The principal objectives of this trial were twofold: (a) to examine the safety and relative efficacy of intradermal needle injection versus s.c. jet administration of a carcinoembryonic antigen (CEA)-encoding recombinant vaccinia virus (rV-CEA) over a limited dose range and (b) to evaluate CEA-specific immune responses or antitumor effects induced by rV-CEA vaccination. Patients were randomly assigned to one of two groups, depending upon the technique of vaccine administration. All 20 patients received two doses of 10(7) or 10(8) pfu of rV-CEA at a 4-week interval. Toxicity was limited to modest local inflammation at the inoculation site as well as low-grade fever and increased fatigue, each affecting fewer than 20% of the patients. No evidence of CEA-specific lymphoproliferation, interleukin 2 release, delayed-type hypersensitivity, or antibody response was observed. Thus, the efficacy comparison between the two administration techniques was based upon the induction of immune responses to the vaccinia virus vector. Both techniques induced vaccinia-specific lymphoproliferation, interleukin 2 release, and antibody responses of comparable magnitude and frequency as well as protected 80% of patients against pustule formation following vaccinia scarification. Thus, there is no compelling reason to recommend one administration technique over the other based upon toxicity or efficacy. We have selected s.c. jet injection for subsequent trials of rV-CEA based on the ability to accommodate larger injection volumes, enhanced standardization between clinicians, and avoidance of needles that could transmit the vaccine or blood-borne pathogens to health care workers. We recommend use of 10(8) pfu doses for subsequent trials of recombinant vaccinia virus vaccines based upon the favorable toxicity profile and more consistent local pustule formation indicative of an adequate inoculation of live virus. No objective clinical responses to the rV-CEA vaccine were observed among this population of patients with widely metastatic adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Adenocarcinoma/therapy , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Vaccinia virus/genetics , Adenocarcinoma/immunology , Adult , Aged , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/immunology , Female , Humans , Injections, Intradermal , Injections, Subcutaneous , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Vaccinia virus/immunology
17.
Gene Ther ; 5(7): 865-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9813656

ABSTRACT

A nucleic acid vaccine encoding human carcinoembryonic antigen (CEA) was administered to 10 juvenile dogs, 10-15 weeks of age, by four parenteral routes. The routes tested were intramuscular injection using a needle and syringe, intramuscular injection using the Biojector needleless injection device, intradermal injection or intravenous injection. All groups received 150 micrograms of plasmid DNA on weeks 0, 4, 7 and 13. All dogs were bled weekly for 17 weeks and tested for antibody against human CEA. Dogs given plasmid intramuscularly either by needle and syringe or Biojector showed significant antibody responses by week 9 which peaked by week 15. Dogs receiving plasmid intravenously showed slight, unsustained increases in antibody titers while dogs receiving plasmid intradermally had significant titers, but at levels approximately one log less than those induced by intramuscular injection. The five dogs immunized by intramuscular delivery of plasmid DNA were examined for cellular immune responses to human CEA by lymphoblast transformation (LBT) assay. All five showed significant CEA-specific lymphoproliferation when compared with unvaccinated dogs. Physical examination, clinical chemistry, hematology and histopathology examinations revealed no abnormalities associated with nucleic acid immunization.


Subject(s)
Antibody Formation , Carcinoembryonic Antigen/genetics , Immunity, Cellular , Vaccines, DNA/administration & dosage , Animals , Antibodies, Monoclonal/blood , Carcinoembryonic Antigen/immunology , Dogs , Humans , Injections, Intradermal , Injections, Intramuscular/instrumentation , Injections, Intramuscular/methods , Injections, Intravenous , Lymphocyte Activation , Neoplasms/therapy
18.
Clin Cancer Res ; 4(11): 2903-12, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829759

ABSTRACT

In preparation for a Phase I trial of DNA immunization against carcinoembryonic antigen (CEA) in patients with colorectal carcinoma, we have produced a single plasmid DNA encoding CEA and hepatitis B surface antigen (HBsAg) under transcriptional regulatory control of two separate cytomegalovirus promoters within separate eukaryotic expression cassettes, designated pCEA/HBsAg. Hepatitis B surface antigen was included to provide an internal positive control for the efficacy of this immunization strategy without regard to the issue of breaking tolerance to a self-antigen. In the present work, we sought to examine the immunogenicity of this plasmid in a nonhuman primate model with close phylogenetic relationship to humans. Groups of pig-tailed macaques were immunized with pCEA/ HBsAg by i.m. injection or particle bombardment of the skin according to a dose and schedule thought to be optimal for the respective technique of DNA immunization. Both administration techniques produced humoral and lympho-proliferative responses of comparable magnitude. However, delayed type hypersensitivity to CEA and CEA-specific interleukin-2 release were observed only in the i.m. group, suggesting a qualitative difference in the character of the immune response elicited by the two techniques of DNA immunization. The antibody responses to CEA and HBsAg were surprisingly persistent in that all immunized animals maintained moderate antibody titers against both antigens for more than 15 months after the last boost. No toxicity was observed during 2 years of follow-up, including no measurable levels of anti-DNA antibody. This antitumor immunization strategy is presently being examined in patients with metastatic colorectal carcinoma using pCEA/HBsAg administered by i.m. injection.


Subject(s)
Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/immunology , Polynucleotides/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Antibodies/blood , Antibodies/immunology , Antibody Formation/drug effects , Cancer Vaccines/immunology , Cancer Vaccines/toxicity , Female , Hepatitis B Surface Antigens/immunology , Immunity, Cellular/drug effects , Immunization , Interleukin-2/metabolism , Lymphocyte Activation , Macaca nemestrina , Polynucleotides/immunology , Polynucleotides/toxicity , Vaccines, DNA/immunology , Vaccines, DNA/toxicity
19.
Immunopharmacology ; 40(1): 1-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9776473

ABSTRACT

The antiproliferative effect of BCX-34 was tested in normal human peripheral blood mononuclear cells (PBMCs) induced to proliferate with OKT3, tetanus toxoid, the mixed lymphocyte reaction, or IL-2. In the case of OKT3, tetanus toxoid, or the MLR the IC50s ranged between 0.7 and 4 microM. With IL-2, the IC50 was 14.6 microM. In T-cells purified by rosetting the IC50 with IL-2 was 0.62 microM. In CD4 or CD8 cells obtained by magnetic activated cell sorting the IC50s with IL-2 were 0.24 and 0.62 microM, respectively. BCX-34 inhibition of proliferation in human PBMCs may not depend entirely upon the accumulation of intracellular dGTP because tetanus toxoid-induced proliferation was inhibited in the absence of deoxyguanosine and was not reversed by deoxycytidine. BCX-34 did not inhibit IL-2 release from PBMCs and did not alter PBMC viability. The results of these studies show that BCX-34 is a potent inhibitor of normal human T-cell proliferation induced by antigenic or IL-2 stimulation. BCX-34 in normal human T-cells has a deoxyguanosine-independent mechanism to suppress in vitro proliferation. BCX-34 appears to have little effect on T-cell viability. The data suggest that BCX-34 may be useful in the treatment of T-cell proliferative disorders.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Immunosuppressive Agents/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , T-Lymphocytes/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Guanine/pharmacology , Humans , Interleukin-2/metabolism , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Muromonab-CD3/pharmacology , T-Lymphocytes/immunology , Tetanus Toxin/pharmacology
20.
Gene Ther ; 4(6): 624-7, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9231080

ABSTRACT

Polynucleotide immunization has been employed as a means of inducing immune responses through the introduction of antigen-encoding DNA. While immunization against specific tumor antigens may be achieved through this strategy, various candidate tumor antigens may not be approached via DNA-based vaccines as they represent transforming oncogenes. As an alternative approach, we have explored the utility of mRNA vectors for polynucleotide immunization. The transient expression achieved by mRNA may provide an efficient and safe system for stimulating immune responses to tumor-specific antigens. Our previous work demonstrated that a self-replicating RNA enhances the magnitude and duration of transgene expression for this application. Here we further modify the vector for optimal use in gene therapy through the incorporation of untranslated regions flanking the encoded transgene. The beta-globin 5' and 3' untranslated regions (UTRs) were inserted directly flanking the luciferase gene in both nonreplicative and replicative RNA constructs. In both cases, elevated and prolonged levels of luciferase expression were detected from the beta-globin UTR-flanked luciferase as compared to luciferase without these sequences. These modifications improve the ability of replicative RNA vectors to produce high, yet transient transgene expression for cancer immunotherapy strategies.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Globins/genetics , Neoplasms/therapy , RNA, Messenger/genetics , Sindbis Virus/genetics , Animals , Antigens, Neoplasm , Cancer Vaccines , Gene Expression , Gene Transfer Techniques , Luciferases/genetics , Mice , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...