Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-36896021

ABSTRACT

Hoefges et al. utilized a whole-proteome peptide array approach to show that C57BL/6 mice develop a large repertoire of antibodies against linear peptide sequences of their melanoma after receiving a curative immunotherapy regimen consisting of radiation and an immunocytokine. Antibodies can play an important role in innate and adaptive immune responses against cancer, and in preventing infectious disease. Flow cytometry analysis of sera of immune mice that were previously cured of their melanoma through a combined immunotherapy regimen with long-term memory showed strong antibody-binding against melanoma tumor cell lines. Using a high-density whole-proteome peptide array, we assessed potential protein-targets for antibodies found in immune sera. Sera from 6 of these cured mice were analyzed with this high-density, whole-proteome peptide array to determine specific antibody-binding sites and their linear peptide sequence. We identified thousands of peptides that were targeted by 2 or more of these 6 mice and exhibited strong antibody binding only by immune, not naive sera. Confirmatory studies were done to validate these results using 2 separate ELISA-based systems. To the best of our knowledge, this is the first study of the "immunome" of protein-based epitopes that are recognized by immune sera from mice cured of cancer via immunotherapy.

2.
J Clin Orthop Trauma ; 8(Suppl 2): S40-S42, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29339843

ABSTRACT

Peri-implant fractures of the wrist are uncommon, and usually present as stress fractures distal to the site of the implant. We report an unusual case where the radius has fractured beneath a plate, causing bending and deformity of the implant. This prevented reduction of the fracture under sedation, so urgent intervention became necessary due to neurovascular compromise.

3.
Biotechnol Lett ; 34(1): 91-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21956129

ABSTRACT

Cellulose degradation is essential for the future production of many advanced biofuels. Cellulases from the filamentous fungus Trichoderma reesei are among the most efficient enzymes for the hydrolysis of cellulosic materials. One of the cellulases from T. reesei, cellobiohydrolase II (CBH2), was studied because of its industrial relevance and proven enzymatic activity. Using both crude and rigorous membrane fractionation methods we show that full length T. reesei CBH2 is exclusively localized to the outer membrane when expressed recombinantly in Escherichia coli. Even fusing signal sequence-free maltose-binding protein to the N-terminus of CBH2, which has been shown to increase solubility of other proteins, did not prevent the outer membrane localization of CBH2. These results highlight the difficulties in producing fungal cellulases in bacterial hosts and provide a stepping stone for future cellulase engineering efforts.


Subject(s)
Bacterial Outer Membrane Proteins/analysis , Cell Membrane/chemistry , Cellulose 1,4-beta-Cellobiosidase/analysis , Escherichia coli/chemistry , Trichoderma/enzymology , Bacterial Outer Membrane Proteins/genetics , Cell Fractionation , Cellulose 1,4-beta-Cellobiosidase/genetics , Cloning, Molecular , Escherichia coli/genetics , Gene Expression , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Trichoderma/genetics
4.
Cancer Gene Ther ; 14(8): 717-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17557109

ABSTRACT

Effective eradication of established tumor and generation of a lasting systemic immune response are the goals of cancer immunotherapy. The objective of this phase IB study was to assess the safety and toxicity of treatment to metastatic tumor underlying the skin with the DNA encoding interleukin-12 (IL-12). This treatment strategy allowed the patient's own tumor to serve as a source of autologous antigen in the tumor microenvironment. We proposed that IL-12 protein produced by the transfected cells would result in the generation of both a local and systemic antitumor response. The tumor was treated with either three or six intratumoral injections of plasmid containing IL-12 DNA. This treatment strategy resulted in no significant local or systemic toxicity. The treatment did not result in an increase in serum IL-12 protein. The size of the treated lesion decreased significantly (greater than 30%) in five of the 12 patients. However, nontreated subcutaneous lesions or other disease did not decrease in size.


Subject(s)
DNA, Neoplasm/administration & dosage , Genetic Vectors/administration & dosage , Interleukin-12/administration & dosage , Interleukin-12/genetics , Melanoma/therapy , Plasmids/genetics , Adult , Aged , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , DNA, Neoplasm/adverse effects , Female , Genetic Vectors/adverse effects , Humans , Injections, Intralesional , Interleukin-12/adverse effects , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Male , Melanoma/genetics , Melanoma/secondary , Middle Aged
6.
J Child Neurol ; 19(3): 231-3, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15119487

ABSTRACT

Two cases of hemorrhagic meningoencephalitis secondary to Naegleria fowleri infection confirmed by postmortem analysis are described. The first patient is a 5-year-old boy who presented with a severe headache, neck stiffness, and lethargy. His neurologic examination was significant for somnolence and nuchal rigidity. Cerebrospinal fluid studies and structural neuroimaging were consistent with hemorrhagic meningoencephalitis. Another 5-year-old boy presented to a different institution 2 miles away in the same week with similar complaints. Both patients declined rapidly and expired within 48 hours of admission secondary to transtentorial herniation caused by the mass effect of inflammation, edema, and hemorrhage with displacement of the brain stem. Histopathologic and immunochemistry analysis of brain tissue revealed the presence of Naegleria trophozoites in both cases.


Subject(s)
Amebiasis/pathology , Central Nervous System Protozoal Infections/pathology , Cerebral Hemorrhage/pathology , Naegleria fowleri , Animals , Brain/pathology , Brain Edema/pathology , Brain Stem/pathology , Child, Preschool , Diagnosis, Differential , Encephalocele/pathology , Fatal Outcome , Humans , Male , Neurologic Examination
7.
Mt Sinai J Med ; 70(6): 393-400, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14647874

ABSTRACT

Aortoiliac and peripheral vascular disease that cannot be treated medically is generally treated by vascular surgeons and/or interventional radiologists. Advances in technology have allowed endovascular therapy to become a therapeutic option for atherosclerotic disease from the aorta to the femoropopliteal region. Distal peripheral disease is still approached surgically; however, further technological advances may lead to a more active role for endovascular therapy at this level.


Subject(s)
Aorta, Abdominal/surgery , Arteriosclerosis/therapy , Popliteal Artery/surgery , Angioplasty, Balloon/methods , Clinical Trials as Topic , Constriction, Pathologic/therapy , Humans , Ischemia/therapy , Leg/blood supply
8.
Biotechniques ; 35(3): 502-4, 506-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14513555

ABSTRACT

T7-based RNA amplification is applied when there is insufficient RNA. The overall extent of amplification can be measured spectrophotometrically (i.e., quantifying RNA yields), but this measurement does not provide information about the RNA amplification of individual genes. Here we describe a method applying real-time quantitative PCR, which enables the monitoring of RNA amplification of individual genes. The amount of RNA before and after T7-based RNA amplification was determined by real-time quantitative PCR for three housekeeping genes: beta-2-microglobulin, porphobilinogen deaminase, and serine dehydratase, which are, respectively, a high, intermediate/low, and low copy transcript. Real-time quantitative PCR appeared to be suitable to determine the extent of RNA amplification, as was reflected by the low intra- and inter-run coefficients of variation of cycle threshold of 1.1%-2.1%. The application of real-time quantitative PCR showed that T7-based RNA amplification is reproducible but might introduce a sequence-specific bias. Real-time quantitative PCR is a novel approach to monitor RNA amplification and is particularly suited to study RNA amplification of individual genes.


Subject(s)
Bacteriophage T7/genetics , Bacteriophage T7/metabolism , Gene Expression Profiling/methods , Nucleic Acid Amplification Techniques/methods , Online Systems , Polymerase Chain Reaction/methods , RNA/genetics , RNA/metabolism
9.
Proteomics ; 3(4): 479-95, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12687615

ABSTRACT

Many aspects of the toxicity of xenobiotic compounds have been attributed to the consequences of covalent modification of specific proteins, but the nature and specificity of protein targets for classes of electrophilic toxins remain largely uncharacterized. For inhaled toxicants, the point of exposure or absorption lies with epithelial cells lining the pulmonary tree. In this study, abundant proteins in human bronchial epithelial cells that are arylated in vitro by two quinonoid compounds, 1,4-benzoquinone (BQ) and 1,4-naphthoquinone (NQ) have been detected using (14)C-labeled quinones and two-dimensional gel electrophoresis. These proteins were identified using matrix assisted laser desorption/ionization mass spectrometry for tryptic mass mapping followed by sequence database searching. Corroborative identification of protein targets was obtained from the apparent isoelectric points, molecular weights, and the use of antibody probes. There were subtle differences in the protein targets of BQ and NQ, but both associated with the following abundant proteins, nucleophosmin, galectin-1, probable protein disulfide isomerase, protein disulfide isomerase, 60 kDa heat shock protein, mitochondrial stress-70 protein, epithelial cell marker protein, and S100-type calcium binding protein A14. We further delineate the properties of these proteins that make them preferred targets and the evidence these adducts present for delivery of these quinones to subcellular compartments.


Subject(s)
Benzoquinones/pharmacology , Bronchi/drug effects , Bronchi/metabolism , Epithelial Cells/drug effects , Naphthoquinones/pharmacology , Proteins/metabolism , Adolescent , Adult , Amino Acid Sequence , Autoradiography , Bronchi/cytology , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Epithelial Cells/metabolism , Female , Humans , Male , Mass Spectrometry , Oxidation-Reduction , Peptide Mapping , Proteins/chemistry
10.
Hum Gene Ther ; 13(14): 1711-21, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12396624

ABSTRACT

The primary objective of this phase I study was to determine the safety of an autologous tumor vaccine given by intradermal injection of lethally irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected autologous melanoma and sarcoma cells. Secondary objectives included validation of the gene delivery technology (particle-mediated gene transfer), determining the host immune response to the tumor after vaccination, and monitoring patients for evidence of antitumor response. Sixteen patients were treated with either of two different doses of GM-CSF-treated tumor cells. One patient received treatment with both doses of tumor cells. No treatment-related local or systemic toxicity was noted in any patient. Patients administered 100% treated cells (i.e., with a preparation of tumor cells that had all been exposed to GM-CSF DNA transfection) had a more extensive lymphocytic infiltrate at the vaccine site than did patients given 10% treated cells (a preparation of tumor cells in which 10% had been exposed to GM-CSF transfection) or nontreated tumor. The generation of a systemic immune response to autologous tumor by a delayed-type hypersensitivity response to the intradermal placement of nontransfected tumor cells was noted in one patient. One patient had a transient partial response of metastatic tumor sites. The entire procedure, from tumor removal to vaccine placement, was accomplished in less than 6 hr in all patients. Four of 17 patient tumor preparations produced greater than 3.0 ng of GM-CSF per 10(6) cells per 24 hr in vitro. The one patient with greater than 30 ng of GM-CSF per 10(6) cells per 24 hr in vitro had positive DTH, a significant histologic inflammatory response, and clinically stable disease. This technique of gene transfer was safe and feasible, but resulted in clinically relevant levels of gene expression in only a minority of patients.


Subject(s)
Biolistics , Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunologic Factors/genetics , Melanoma/therapy , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Vaccines, DNA/therapeutic use , Cancer Vaccines/administration & dosage , DNA, Complementary/genetics , Dose-Response Relationship, Immunologic , Feasibility Studies , Gold , Humans , Hypersensitivity, Delayed/immunology , Injections, Intradermal , Leiomyosarcoma/pathology , Leiomyosarcoma/therapy , Liposarcoma/pathology , Liposarcoma/therapy , Melanoma/pathology , Melanoma/secondary , Safety , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Treatment Outcome , Tumor Cells, Cultured/transplantation , Vaccines, DNA/administration & dosage
11.
Hematol Oncol Clin North Am ; 15(4): 703-21, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11676280

ABSTRACT

Preclinical and clinical development of antitumor strategies using mAbs are showing antitumor efficacy in animal models and in some clinical settings. Preclinical models suggest that mAb treatment would be most effective when provided in the minimal residual disease setting and can involve mAbs in a variety of roles. In murine models, the combination of mAbs with recombinant cytokines, such as IL-2, IL-12, or GM-CSF, can augment the immunologic effect of the mAbs by activating effector cell functions. Efficacy appears to be greatest when the mAb can recruit the effector cells of the host's immune system into helping in the mediation of the antitumor effect. Clinical testing of these concepts is under way.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Neoplasms/therapy , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Apoptosis , Bone Marrow Purging , Cancer Vaccines/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Complement Activation , Cytokines/therapeutic use , Drug Design , Genetic Engineering , Humans , Interleukin-2/genetics , Interleukin-2/therapeutic use , Lymphocyte Activation , Mice , Molecular Mimicry , Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
12.
Cancer ; 88(8): 1892-901, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10760767

ABSTRACT

BACKGROUND: Interleukin-2 (IL-2) and granulocyte-macrophage-colony stimulating factor (GM-CSF) are cytokines with nonoverlapping pleiotropic effects. In a prior Phase Ib study, this combination of agents exhibited antitumor effects in the lungs of four of eight patients with renal cell carcinoma and pulmonary metastases. We conducted this Phase Ib/II trial to determine the response rate of renal cell carcinoma patients with pulmonary metastases treated with continuous infusion IL-2 plus GM-CSF. METHODS: Patients with renal cell carcinoma and pulmonary metastases were treated with 1.5, 2.25, or 4.5 x 10(6) IU/m(2)/day 96-hour continuous infusion IL-2 on Days 1-4, 8-11, and 15-18, and 1.25, 2.25, or 2.5 microg/kg/day GM-CSF on Days 8-19. RESULTS: Sixteen patients were treated per protocol, 14 of whom could be evaluated for disease progression. None of these 14 patients had >50% shrinkage of either total tumor burden or pulmonary metastasis. One patient developed Grade 5 neurotoxicity. Autopsy revealed acute multifocal cerebral venous thrombosis as well as acute subdural and subarachnoid hemorrhage. CONCLUSIONS: The combination of IL-2 and GM-CSF may be associated with marked morbidity and, as in one case in this study, mortality. No significant antitumor activity was appreciated. Thus, the combination of IL-2 and GM-CSF, when administered at this dose and according to this schedule, does not appear to be active in renal cell carcinoma and is associated with significant toxicities. Further studies using this combination of agents should only be undertaken with extreme caution and particular attention to neurotoxicity.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/secondary , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Adult , Aged , Cerebral Hemorrhage/chemically induced , Drug Administration Schedule , Drug Therapy, Combination , Fatal Outcome , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Interleukin-2/adverse effects , Intracranial Thrombosis/chemically induced , Male , Middle Aged , Nervous System/drug effects , Treatment Outcome , Venous Thrombosis/chemically induced
13.
Cancer Immunol Immunother ; 48(5): 219-29, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10478638

ABSTRACT

The fusion protein formed from ch14.18 and interleukin-2 (ch14.18-IL-2), shown to exhibit antitumor efficacy in mouse models, consists of IL-2 genetically linked to each heavy chain of the ch14.18 chimeric anti-GD2 monoclonal antibody. The purpose of this study was to determine the pharmacokinetics of ch14.18-IL-2 in mice and assess its stability in murine serum. Following i.v. injection, the fusion protein was found to have a terminal half-life of 4.1 h. Detection of IL-2 following injection of the ch14.18-IL-2 fusion protein showed a similar half-life, indicating that the fusion protein prolongs the circulatory half-life of IL-2. Detection of human IgG1 following injection of ch14.18-IL-2 showed a terminal half-life of 26.9 h. These data suggested that the native fusion protein is being altered in vivo, resulting in a somewhat rapid loss of detectable IL-2, despite prolonged circulation of its immunoglobulin components. In vitro incubation of the ch14.18-IL-2 fusion protein in pooled mouse serum at 37 degrees C for 48 h resulted in a loss of its IL-2 component, as detected in enzyme-linked immunosorbent assay systems and in proliferation assays. Polyacrylamide gel electrophoresis and Western blot analysis of the fusion protein incubated in mouse serum at 37 degrees C indicated that the ch14.18-IL-2 is cleaved, resulting in a loss of the 67-kDa band (representing the IL-2 linked to the IgG1 heavy chain) and the detection of a band of more than 50 kDa, slightly heavier than the IgG1 heavy chain itself. This suggests that the fusion protein is being cleaved in vitro within the IL-2 portion of the molecule. These studies show that (1) ch14.18-IL-2 prolongs the circulatory half-life of IL-2 (compared to that of soluble IL-2) and (2) the in vivo clearance of the fusion protein occurs more rapidly than the clearance of the ch14.18 antibody itself, possibly reflecting in vivo cleavage within the IL-2 portion of the molecule, resulting in loss of IL-2 activity.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Interleukin-2/chemistry , Interleukin-2/pharmacokinetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Drug Stability , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunization, Passive/methods , Interleukin-2/blood , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/blood , Temperature , Time Factors
14.
Clin Diagn Lab Immunol ; 6(2): 236-42, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10066660

ABSTRACT

Preliminary testing has shown in vitro and in vivo that antitumor activity can be obtained with fusion proteins linking tumor-reactive monoclonal antibodies to cytokines, such as granulocyte-macrophage colony-stimulating factor or interleukin 2 (IL-2). Preclinical and clinical testing of these reagents requires their in vitro and in vivo quantitation and pharmacokinetic evaluation. We have focused on the detection of a fusion protein which links one human IL-2 molecule to the carboxy terminus of each heavy chain of the tumor-reactive human-mouse chimeric anti-GD2 antibody, ch14.18. We have developed enzyme-linked immunosorbent assays (ELISAs) to evaluate intact tumor-reactive fusion proteins. By these ELISAs we can reliably measure nanogram quantities of intact ch14.18-IL-2 fusion protein and distinguish the intact protein from its components (ch14.18 and IL-2) in buffer, mouse serum, and human serum with specificity and reproducibility. The measurement of intact ch14.18-IL-2 fusion protein is not confounded by free IL-2 or free ch14.18 when 100 ng or less of total immunoglobulin per ml is used during the assay procedure. Our results indicate that these ELISAs are suitable for preclinical and clinical testing and with slight modifications are applicable to the analysis of a variety of other fusion proteins.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Immunoglobulin G/analysis , Interleukin-2/analysis , Recombinant Fusion Proteins/analysis , Animals , Antibodies, Monoclonal , Binding, Competitive/immunology , Blood Proteins , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay/standards , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunoglobulin G/immunology , Indicator Dilution Techniques , Interleukin-2/immunology , Mice , Recombinant Fusion Proteins/immunology , Reproducibility of Results
15.
Clin Cancer Res ; 5(2): 281-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10037176

ABSTRACT

Interleukin-2 (IL-2) is a potent lymphokine that activates natural killer cells, T cells, and other cells of the immune system. Several distinct recombinant human IL-2 preparations have shown antitumor activity, particularly for renal cell cancer and melanoma. Somewhat distinct immune and clinical effects have been noted when different IL-2 preparations have been tested clinically; however, the regimens and doses used were not identical. To compare these more directly, we have evaluated two clinical recombinant IL-2 preparations in vitro and in vivo using similar regimens and similar IUs of IL-2. We used the Food and Drug Administration-approved, commercially available Chiron IL-2 and the Hoffmann LaRoche (HLR) IL-2 supplied by the National Cancer Institute. Using equivalent IUs of IL-2, we noted quantitative differences in vitro and in vivo in the IL-2 activity of these two preparations. In patients receiving comparable IUs of the two preparations, HLR IL-2 induced the release of more soluble IL-2 receptor alpha into the serum than Chiron IL-2. In addition, more toxicities were noted in patients receiving 1.5 x 10(6) IU of HLR IL-2 than were seen in patients treated with 1.5 x 10(6) or even 4.5 x 10(6) IU of Chiron IL-2. These toxicities included fever, nausea and vomiting, and hepatic toxicity. In vitro proliferative assays using IL-2-dependent human and murine cell lines indicated that the IU of HLR IL-2 was more effective than Chiron IL-2 at inducing tritiated thymidine incorporation. Using flow cytometry, we also found quantitative differences in the ability of these two preparations to bind to IL-2 receptors. These findings indicate that approximately 3-6 IU of Chiron IL-2 are required to induce the same biological effect as 1 IU of HLR IL-2.


Subject(s)
Interleukin-2/therapeutic use , Neoplasms/drug therapy , Animals , CD56 Antigen/immunology , Cell Division/drug effects , Humans , Infusion Pumps , Interleukin-2/adverse effects , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lymphocytosis/chemically induced , Mice , Neoplasms/immunology , Receptors, Interleukin-2/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Reference Standards , Serum Albumin/pharmacology , Tumor Cells, Cultured
17.
Cancer Immunol Immunother ; 46(6): 327-37, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9756417

ABSTRACT

Administration of the cytokine interleukin-2 (IL-2) can result in therapeutic benefits for individuals with renal cell carcinoma and melanoma. Here we report an analysis of the transcription factor families AP-1, Sp1, NF-kappaB, and signal transducers and activators of transcription (STAT) in cancer patients' lymphocytes before and after IL-2 immunotherapy, as assessed by a gel-shift assay. An in vitro surrogate of IL-2 immunotherapy is the incubation of fresh peripheral blood mononuclear cells (PBMC) from healthy individuals in IL-2 for several days, resulting in the production of lymphokine-activated killer (LAK) activity in these cultures. One purpose of this study was to describe the profile of transcription factor activation in these different populations, and assess whether the patterns observed correlated with functional differences in these cells. Prior to in vivo IL-2 administration, the typical binding pattern of transcription factors in PBMC from patients resembled that seen in fresh PBMC from healthy individuals. Over a 3-week course of IL-2 therapy, in most patients the binding patterns of AP-1 , Sp1, and NF-kappaB proteins changed to resemble those seen in PBMC activated by IL-2 in vitro. However, the cells obtained from IL-2-treated patients did not have low-level constitutive expression of STAT binding factors as did LAK cells. When these patient cells were further stimulated by IL-2 in vitro, additional differences in STAT induction patterns were noted. These data provide further information on the molecular events occurring in immune cells generated through in vivo and in vitro administration of IL-2, and further document that there is not a precise congruence between PBMC activated in vivo and in vitro by IL-2.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Immunologic Factors/pharmacology , Immunotherapy , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/drug effects , Lymphocytes/drug effects , Melanoma/therapy , Signal Transduction/drug effects , Transcription Factors/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Immunologic Factors/therapeutic use , Interleukin-2/therapeutic use , Killer Cells, Lymphokine-Activated/metabolism , Lymphocytes/metabolism , Melanoma/immunology , NF-kappa B/metabolism , STAT1 Transcription Factor , STAT2 Transcription Factor , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/classification
18.
Clin Cancer Res ; 4(5): 1287-96, 1998 May.
Article in English | MEDLINE | ID: mdl-9607589

ABSTRACT

Lymphocytes from patients receiving in vivo interleukin (IL)-2 therapy possess enhanced in vitro proliferative and cytotoxic responses to IL-2. The cells from these patients that respond to exogenous IL-2 are CD56+ natural killer cells expressing intermediate-affinity IL-2 receptor betagamma(c) complexes. Because IL-15 activates cells via these same betagamma(c) receptors, we hypothesized that IL-15 would also activate lymphocytes from patients treated with in vivo IL-2 therapy and therefore that IL-15 might potentially be useful as an immunotherapeutic agent alone or in combination with IL-2. We report here that peripheral blood mononuclear cells (PBMCs) from patients receiving in vivo IL-2 therapy do proliferate in response to IL-15. However, a greater dose of IL-15 is needed to reach the same level of proliferation stimulated by IL-2. The EC50 for IL-2 is 0.21 +/- 0.04 nM (mean +/- SE; n = 18), whereas the EC50 for IL-15-stimulated proliferation is 1.16 +/- 0.16 nM (n = 18). In contrast to the proliferative response, equivalent doses of IL-2 and IL-15 stimulate patient PBMCs to mediate similar levels of cytotoxicity against Daudi, K562, and LA-N-5 tumor targets. Notably, low concentrations of IL-15 that do not stimulate a substantial proliferative response (e.g., 1.0 ng/ml) do boost PBMCs to mediate cytotoxicity against these tumor targets. These distinct dose-response curves for proliferation compared to cytotoxicity suggest that IL-15 should be evaluated for its potential as an immunotherapeutic agent to treat cancer, particularly in regimens providing doses that might minimize the proliferative response (associated with cytokine release and toxic side effects) while maintaining the cytolytic antitumor response.


Subject(s)
Interleukin-15/pharmacology , Interleukin-2/pharmacology , Lymphocyte Subsets/drug effects , Melanoma/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD56 Antigen/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Interleukin-2/therapeutic use , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Lectins, C-Type , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Lymphocyte Subsets/cytology , Melanoma/drug therapy , Tumor Cells, Cultured
19.
Clin Cancer Res ; 3(8): 1277-88, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9815810

ABSTRACT

We conducted a Phase IB trial of antidisialoganglioside chimeric 14. 18 (ch14.18) antibody and interleukin 2 (IL-2) to determine the maximal tolerated dose (MTD), immunological effects, antitumor effects, and toxicity of this treatment combination. Twenty-four melanoma patients received immunotherapy with ch14.18 antibody and a continuous infusion of Roche IL-2 (1.5 x 10(6) units/m2/day) given 4 days/week for 3 weeks. The ch14.18 antibody (dose level, 2-10 mg/m2/day) was scheduled to be given for 5 days, before, during, or following initial systemic IL-2 treatment. The ch14.18 MTD was 7.5 mg/m2/day, and 15 patients were treated with the ch14.18 MTD. Immunological effects included the induction of lymphokine-activated killer activity and antibody-dependent cellular cytotoxicity by peripheral blood mononuclear cells. In addition, serum samples obtained following ch14.18 infusions were able to facilitate in vitro antibody-dependent cellular cytotoxicity. Antitumor activity included one complete response, one partial response, eight patients with stable disease, and one patient with >50% decrease of hepatic metastases in the face of recurrence of a s.c. lesion. Dose-limiting toxicities were a severe allergic reaction and weakness, pericardial effusion, and decreased performance status. Most patients treated at the MTD had abdominal, chest, or extremity pain requiring i.v. morphine. One patient had an objective peripheral neuropathy. This IL-2 and ch14.18 treatment combination induces immune activation in all patients and antitumor activity in some melanoma patients. We are attempting to enhance this treatment approach by addition of the anti-GD3 R24 antibody to this IL-2 and ch14.18 regimen.


Subject(s)
Antibodies, Monoclonal/adverse effects , Interleukin-2/adverse effects , Melanoma/therapy , Adult , Antibodies, Anti-Idiotypic/blood , Antibody-Dependent Cell Cytotoxicity , Dose-Response Relationship, Drug , Female , Humans , Immunophenotyping , Immunotherapy , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Count , Lymphocytes/immunology , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Recombinant Fusion Proteins/adverse effects , Recombinant Proteins/adverse effects , Tumor Cells, Cultured
20.
Cancer ; 80(2): 317-33, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9217046

ABSTRACT

BACKGROUND: The murine monoclonal antibody (MoAb) 14.G2a recognizes GD2, a disialoganglioside expressed in tumors of neuroectodermal origin, and facilitates antibody dependent cellular cytotoxicity (ADCC) in vitro. When given in vivo, interleukin-2 (IL-2) can increase ADCC by enhancing the activity and number of circulating lymphocytes. METHODS: Thirty-three pediatric patients with GD2 positive malignancies, ranging in age from 2 to 17 years (median, 9.9 years), received IL-2 and 14.G2a in this Phase I/IB study of the Children's Cancer Group (CCG) and were monitored for toxicities and response to therapy. Seven of these patients also received granulocyte-macrophage-colony stimulating factor. RESULTS: The maximum tolerated dose (MTD) of 14.G2a with IL-2 was 15 mg/m2/day. The most prevalent Grade 3-4 toxicities were generalized pain (n = 14 [42%]) and fever without documented infection (n = 17 [52%]). IL-2 was thought to be the causative agent in most cases of fever. Toxicities attributed to 14.G2a included pain, allergic or anaphylactic reactions, and rash. Human antimouse antibodies were demonstrated in 9 of 21 evaluated patients. One patient with neuroblastoma had a partial response, and one patient with osteosarcoma had a complete response. Immunocytology demonstrated that the number of neuroblastoma cells in bone marrow decreased in three patients. CONCLUSIONS: The murine MoAb 14.G2a was well tolerated at the MTD and appeared to have some antitumor activity. Further development of this approach will involve additional engineered forms of the antibody as well as testing in the adjuvant and minimal residual disease setting.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Gangliosides/immunology , Interleukin-2/therapeutic use , Neoplasm Proteins/immunology , Neuroblastoma/therapy , Adjuvants, Immunologic/adverse effects , Adolescent , Animals , Antibodies, Monoclonal/adverse effects , Antibody-Dependent Cell Cytotoxicity , Bone Marrow/pathology , Child , Child, Preschool , Female , Humans , Interleukin-2/adverse effects , Male , Mice , Neuroblastoma/pathology , Recombinant Proteins/therapeutic use , Remission Induction
SELECTION OF CITATIONS
SEARCH DETAIL
...