Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
PLoS One ; 10(6): e0129954, 2015.
Article in English | MEDLINE | ID: mdl-26091536

ABSTRACT

BACKGROUND: Interleukin (IL)-12 is a pro-inflammatory cytokine that mediates T-helper type 1 responses and cytotoxic T-cell activation, contributing to its utility as anti-cancer agent. Systemic administration of IL-12 often results in unacceptable toxicity; therefore, strategies to direct delivery of IL-12 to tumors are under investigation. The objective of this study was to assist the preclinical development of NHS-IL12, an immunocytokine consisting of an antibody, which targets necrotic tumor regions, linked to IL-12. Specifically this study sought to evaluate the safety, serum pharmacokinetics, anti-tumor activity, and immune modulation of NHS-IL12 in dogs with naturally occurring cancers. METHODOLOGY/PRINCIPAL FINDINGS: A rapid dose-escalation study of NHS-IL12 administered subcutaneously to dogs with melanoma was conducted through the Comparative Oncology Trials Consortium (COTC). Eleven dogs were enrolled in four dose-escalation cohorts; thereafter, an additional seven dogs were treated at the defined tolerable dose of 0.8 mg/m2. The expanded cohort at this fixed dose (ten dogs in total) was accrued for further pharmacokinetics and pharmacodynamics assessment. NHS-IL12 levels, serum cytokine concentrations, and peripheral blood mononuclear cell characterization (post-treatment) and draining lymph node immune profiling, and tumor biopsies (pre- and post-treatment) were collected. Adverse events included thrombocytopenia, liver enzymopathies, fever, and vasculitis. Correlation between interferon (IFN)-γ induction, adverse events, and NHS-IL12 exposure (maximum concentration and area under the concentration-time curve) were dose-dependent. Serum IL-10 levels and intratumoral CD8+ populations increased after treatment. Partial responses, according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, were observed in two dogs treated with NHS-IL12 0.8 mg/m2 and 1.6 mg/m2. CONCLUSIONS/SIGNIFICANCE: NHS-IL12 was administered safely to dogs with melanoma and both immunologic and clinical activity was observed. This study successfully defined a narrow therapeutic window for systemic delivery of NHS-IL12 via the subcutaneous route. Results will inform the design and implementation of first-in-human clinical trials of NHS-IL12 in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Dog Diseases/drug therapy , Dog Diseases/pathology , Immunoglobulin G/pharmacology , Immunologic Factors/pharmacology , Interleukin-12/pharmacology , Melanoma/veterinary , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/blood , Dog Diseases/blood , Dogs , Female , Immunoglobulin G/administration & dosage , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Immunophenotyping , Infusions, Subcutaneous , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
2.
J Exp Clin Cancer Res ; 32: 5, 2013 Jan 26.
Article in English | MEDLINE | ID: mdl-23352035

ABSTRACT

BACKGROUND: An adenovirus that expresses both interleukin (IL)-12 and granulocyte-macrophage colony-stimulating-factor (GM-CSF) has been proven to be very effective in treating several tumors, but causes serious normal tissue toxicities. METHODS: In this study, a novel adenoviral vector was constructed by placing the human GM-CSF gene under the control of the CMV-IE promoter and human IL-12 gene under the control of heat shock protein 70B gene promoter. Both hGM-CSF and hIL-12 expressions in virus-infected tumor cells were analyzed in vitro and in vivo when underlying single or multiple rounds of hyperthermia. RESULTS: We observed constitutive high expression of human GM-CSF and heat-induced expression of human IL-12 after a single round of hyperthermia post viral infection. The heat-induced hIL-12 expression exhibited a pulse-like pattern with a peak at 24 hrs followed by a decline 48 hrs post heat stress. Repeated heat treatment was more effective in inducing hIL-12 expression than a one-time heat treatment. Interestedly, we also observed that constitutive expression of hGM-CSF could be stimulated by heat stress in tested tumor cells. CONCLUSION: Our study provided a novel strategy for combined gene therapy that allows constitutive expression of a non-toxic gene such as GM-CSF and heat-induced expression of a toxic gene such as IL-12. In addition, our study also showed that hyperthermia can be used to trigger gene expression in temporal and special manner.


Subject(s)
Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hyperthermia, Induced/methods , Interleukin-12/genetics , Adenoviridae/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/virology , Cell Culture Techniques , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Humans , Interleukin-12/biosynthesis , Interleukin-12/pharmacokinetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Liver Neoplasms/virology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Lung Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 17(7): 1998-2005, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21447719

ABSTRACT

PURPOSE: AS1409 is a fusion protein comprising a humanized antibody BC1 linked to interleukin-12 (IL-12). It is designed to deliver IL-12 to tumor-associated vasculature using an antibody targeting the ED-B variant of fibronectin. EXPERIMENTAL DESIGN: We conducted a phase 1 trial of weekly infusional AS1409 in renal carcinoma and malignant melanoma patients. Safety, efficacy, markers of IL-12-mediated immune response, and pharmacokinetics were evaluated. RESULTS: A total of 11 melanoma and 2 renal cell carcinoma patients were treated. Doses of 15 and 25 µg/kg were studied. Most drug-related adverse events were grade 2 or less, and included pyrexia, fatigue, chills, headache, vomiting, and transient liver function abnormalities. Three dose limiting toxicities of grade 3 fatigue and transaminase elevation were seen at 25 µg/kg. IFN-γ and interferon-inducible protein-10 (IP-10) were elevated in all patients, indicating activation of cell-mediated immune response; this was attenuated at subsequent cycles. Antidrug antibody responses were seen in all patients, although bioassays indicate these do not neutralize AS1409 activity. Plasma half-life was 22 hours and not dose-dependent. Five patients received 6 cycles or more and a best response of at least stable disease was seen in 6 (46%) patients. Partial response was seen in a melanoma patient, and disease shrinkage associated with metabolic response was maintained beyond 12 months in another melanoma patient despite previous rapid progression. CONCLUSIONS: The maximum tolerated dose was established at 15 µg/kg weekly. AS1409 is well tolerated at this dose. Evidence of efficacy assessed by RECIST, functional imaging, and biomarker response warrants the planned further investigation using this dose and schedule in malignant melanoma.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Interleukin-12/therapeutic use , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Adult , Aged , Antibodies/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacokinetics , Carcinoma, Renal Cell/immunology , Female , Humans , Interferon-gamma/blood , Interleukin-12/genetics , Interleukin-12/pharmacokinetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome , Tumor Burden/drug effects , Young Adult
4.
Pharmacology ; 85(6): 319-27, 2010.
Article in English | MEDLINE | ID: mdl-20516733

ABSTRACT

BACKGROUND/AIMS: Interleukin-12 (IL-12) is a cytokine that plays an important role in cell-mediated immunity and shows great potential as a therapeutic agent for the treatment of tumors and infectious diseases. METHODS: We investigated the pharmacokinetics (PK) and pharmacodynamics of recombinant human IL-12 (rhIL-12) and rhIL-12 combined with hepatitis B surface antigen (HB(s)Ag) after administration by subcutaneous (s.c.) injection or intravenous infusion in cynomolgus monkeys. RESULTS: After s.c. injection of rhIL-12 at doses of 0.15-1.5 microg/kg, the monkey's metabolism showed linear kinetic characteristics. The intramuscular injection of HB(s)Ag vaccine did not affect the pharmacokinetic profile of rhIL-12. In monkeys administered rhIL-12 in a continuous dosing fashion, serum rhIL-12 was undetectable, probably due to the neutralizing effect of anti-rhIL-12 antibodies. In monkeys receiving high-dose s.c. injection of rhIL-12, the T(max) for serum rhIL-12 concentration was 4-8 h, and the T(max) for serum interferon-gamma (IFN-gamma) concentration was 24-72 h. However, in monkeys receiving continuous dosing of rhIL-12, serum IFN-gamma concentration was very low or even undetectable. CONCLUSION: We found that the PK of rhIL-12 was dose-dependent and its pharmacological effects appeared after T(max) and lasted much longer than mean retention time.


Subject(s)
Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Interleukin-12/pharmacology , Interleukin-12/pharmacokinetics , Adjuvants, Immunologic , Animals , Female , Hepatitis B Surface Antigens/blood , Hepatitis B Vaccines/immunology , Humans , Infusions, Intravenous , Injections, Subcutaneous , Interleukin-12/administration & dosage , Interleukin-12/blood , Macaca fascicularis , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
5.
J Orthop Res ; 28(1): 48-54, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19588527

ABSTRACT

The increasing incidence of bacterial infection and the appearance of Staphylococcus aureus (S. aureus) strains that are resistant to commonly used antibiotics has made it important to develop non-antibiotic approaches for infection prevention. The aim of this study was to develop local monocyte chemoattractant protein-1 (MCP-1) and interleukin-12 p70 (IL-12 p70) therapies to prevent S. aureus infection by enhancing the recruitment and activation of macrophages, which are believed to play an important role in infection prevention as the first line of defense against invading pathogens. Nanocoating systems for MCP-1 and IL-12 p70 deliveries were prepared, and their release characteristics desirable for infection prevention in open fractures were explored. Local MCP-1 therapy reduced S. aureus infection and influenced white blood cell populations, and local IL-12 p70 treatment had a more profound effect on preventing S. aureus infection. No synergistic relationship in decreasing S. aureus infection was observed when MCP-1 and IL-12 p70 treatments were combined. This reported new approach may reduce antibiotic use and antibiotic resistance.


Subject(s)
Chemokine CCL2/administration & dosage , Coated Materials, Biocompatible/administration & dosage , Fractures, Open/surgery , Nanostructures , Osteomyelitis/prevention & control , Peptide Fragments/administration & dosage , Prosthesis-Related Infections/prevention & control , Staphylococcal Infections/prevention & control , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Animals , Bone Wires/adverse effects , Chemokine CCL2/pharmacokinetics , Coated Materials, Biocompatible/pharmacokinetics , Disease Models, Animal , Drug Resistance, Bacterial , Femoral Fractures/complications , Femoral Fractures/surgery , Fracture Fixation, Intramedullary/adverse effects , Fracture Fixation, Intramedullary/instrumentation , Fractures, Open/complications , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Internal Fixators/adverse effects , Male , Nanotechnology/methods , Osteomyelitis/microbiology , Peptide Fragments/pharmacokinetics , Prosthesis Design , Prosthesis-Related Infections/microbiology , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects
6.
Biochem Biophys Res Commun ; 367(2): 330-5, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18158918

ABSTRACT

For a valid cytokine immunotherapy of malignancies, a suitable delivery system that ensures slow-release of cytokines is required, because short half-life in vivo of the molecules ruins therapeutic efficacy while causing severe systemic toxic effects. We previously showed that the cholesterol-bearing pullulan (CHP)-based hydrogel nanoparticles, or nanogel, encapsulates, stabilizes and releases various molecules. Here we applied this nanogel to administration in vivo of interleukin-12 (IL-12). Recombinant murine IL-12 (rmIL-12) was successfully incorporated into CHP nanogel simply by incubated with CHP at room temperature. After subcutaneously injected into mice, the CHP/rmIL-12 complex led to a prolonged elevation in IL-12 concentration in the sera. Repetitive administrations of the CHP/rmIL-12, but not rmIL-12 alone, induced drastic growth retardation of preestablished subcutaneous fibrosarcoma without causing any serious toxic event. The present study proposes a novel therapeutic intervention technology, taking advantage of slow and sustained release of bioactive cytokines from the self-assembling biocompatible nanoparticles.


Subject(s)
Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Nanostructures/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cholesterol/chemistry , Fibrosarcoma/pathology , Gels/chemistry , Glucans/chemistry , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanostructures/administration & dosage , Treatment Outcome
7.
Cancer Immunol Immunother ; 56(4): 447-57, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16874486

ABSTRACT

IL-12 is a cytokine which showed anti-tumor effects in clinical trials, but also produced serious toxicity. We describe a fusion protein, huBC1-IL12, designed to achieve an improved therapeutic index by specifically targeting IL-12 to tumor and tumor vasculature. huBC-1 is a humanized antibody that targets a cryptic sequence of the human ED-B-containing fibronectin isoform, B-FN, present in the subendothelial extracellular matrix of most aggressive tumors. B-FN is oncofetal and angiogenesis-associated, and is undetectable in most normal adult tissues. The original murine BC-1 antibody has been used successfully for immunoscintigraphy to image brain tumor mass in glioblastoma patients. In huBC1-IL12, each of the IgG heavy chains is genetically fused to the N-terminus of the IL-12 p35 subunit, which in turn is disulfide-bonded to the p40 subunit, resulting in a hexameric molecule of MW of approximately 300 kDa. Since human IL-12 has no biological activity in mice, we produced huBC1-muIL12 as a surrogate molecule for animal tumor models. Despite the relatively poor PK profile of this molecule in mice and the apparent drawbacks of xenogeneic models in SCID mice, which lack T and B cells, one cycle of treatment with huBC1-muIL12 was efficacious in the PC3mm2, A431, and HT29 subcutaneous tumor models and PC3mm2 lung metastasis model. This molecule also was found to have surprisingly low toxicity in immunocompetent mice. A fusion protein that contains human IL-12 (huBC1-huIL12), which is a suitable molecule for investigation as a therapeutic, has also been produced. This protein has been shown to have a longer serum half-life than huBC1-muIL12 in mice, and retains both antigen binding and IL-12 activity in in vitro assays.


Subject(s)
Fibronectins/immunology , Interleukin-12/therapeutic use , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/therapeutic use , Animals , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunoglobulin G , Immunoglobulin Heavy Chains , Immunohistochemistry , Interleukin-12/immunology , Interleukin-12/pharmacokinetics , Male , Mice , Neoplasms, Experimental/immunology , Protein Isoforms/immunology , Protein Isoforms/pharmacokinetics , Protein Isoforms/therapeutic use , Recombinant Fusion Proteins/immunology
8.
Clin Cancer Res ; 10(8): 2626-35, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15102664

ABSTRACT

The objective of this Phase II study was to evaluate the pharmacodynamic and immune effects of intratumorally administered recombinant human interleukin-12 (IL-12) on regional lymph nodes, primary tumor, and peripheral blood. Ten previously untreated patients with head and neck squamous cell carcinoma were injected in the primary tumor two to three times, once/week, at two dose levels of 100 or 300 ng/kg, before surgery. We compared these patients with 20 control (non-IL-12-treated) patients. Toxicity was high, with unexpected dose-limiting toxicities at the 300 ng/kg dose level. Dose-dependent plasma IFN-gamma and IL-10 increments were detected. These cytokine levels were higher after the first injection than after the subsequent injections. A rapid, transient reduction in lymphocytes, monocytes, and all lymphocyte subsets, especially natural killer cells, was observed, due to a redistribution to the lymph nodes. In the enlarged lymph nodes of the IL-12-treated patients, a higher percentage of natural killer cells and a lower percentage of T-helper cells were found compared with control patients. The same pattern was detected in the infiltrate in the primary tumor. Real-time semiquantitative PCR analysis of peripheral blood mononuclear cells in the peripheral blood showed a transient decrease of T-bet mRNA. Interestingly, the peripheral blood mononuclear cells in the lymph nodes showed a 128-fold (mean) increase of IFN-gamma mRNA. A switch from the Th2 to a Th1 profile in the lymph nodes compared with the peripheral blood occurred in the IL-12-treated patients. In conclusion, in previously untreated head and neck squamous cell carcinoma patients, recombinant human IL-12 intratumorally showed dose-limiting toxicities at the dose level of 300 ng/kg and resulted in measurable immunological responses locoregionally at both dose levels.


Subject(s)
Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Lymph Nodes/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Th1 Cells/cytology , Adult , Aged , Area Under Curve , Case-Control Studies , Cytokines/blood , Female , Humans , Interferon-gamma/blood , Interleukin-10/blood , Kinetics , Leukocytes, Mononuclear/metabolism , Lymph Nodes/drug effects , Lymphocyte Subsets/drug effects , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Recombinant Proteins/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Outcome
9.
J Immunol ; 172(9): 5159-67, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100252

ABSTRACT

Interleukin-12 can act as a potent adjuvant for T cell vaccines, but its clinical use is limited by toxicity. Paracrine administration of IL-12 could significantly enhance the response to such vaccines without the toxicity associated with systemic administration. We have developed a novel vaccine delivery system (designated F2 gel matrix) composed of poly-N-acetyl glucosamine that has the dual properties of a sustained-release delivery system and a potent adjuvant. To test the efficacy of paracrine IL-12, we incorporated this cytokine into F2 gel matrix and monitored the response of OT-1 T cells in an adoptive transfer model. Recipient mice were vaccinated with F2 gel/SIINFEKL, F2 gel/SIINFEKL/IL-12 (paracrine IL-12), or F2 gel/SIINFEKL plus systemic IL-12 (systemic IL-12). Systemic levels of IL-12 were lower in paracrine IL-12-treated mice, suggesting that paracrine administration of IL-12 may be associated with less toxicity. However, paracrine administration of IL-12 was associated with an enhanced Ag-specific T cell proliferative and functional response. Furthermore, paracrine IL-12 promoted the generation of a stable, functional memory T cell population and was associated with protection from tumor challenge. To study the mechanisms underlying this enhanced response, wild-type and gene-deficient mice were used. The enhanced immune response was significantly reduced in IFN-gamma(-/-) and IL-12R beta 2(-/-) recipient mice suggesting that the role of IL-12 is mediated, at least in part, by host cells. Collectively, the results support the potential of F2 gel matrix as a vaccine delivery system and suggest that sustained paracrine release of IL-12 has potential clinical application.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Interferon-gamma/biosynthesis , Interleukin-12/administration & dosage , Vaccines, Subunit/immunology , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/physiology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Egg Proteins/administration & dosage , Egg Proteins/immunology , Epitopes, T-Lymphocyte/administration & dosage , Gels , Immunologic Memory , Interferon-gamma/physiology , Interleukin-12/metabolism , Interleukin-12/pharmacokinetics , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Paracrine Communication/immunology , Peptide Fragments , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-12 , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Vaccines, Subunit/administration & dosage
10.
Cancer Biother Radiopharm ; 19(6): 764-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15665625

ABSTRACT

Controlled-release, injectable polymer microspheres provide a clinically feasible alternative to gene-modification for the local, sustained delivery of cytokines to tumors for cancer immunotherapy. Long-term release kinetics, bioactivity profiles, and stability of interleukin-2 (IL-2), interleukin-12 (IL-12), and granulocyte- macrophage colony-stimulating factor (GM-CSF)-encapsulated microspheres prepared by phase inversion nanoencapsulation (PIN) were evaluated. While all formulations released physiologically relevant quantities of cytokine for up to 30 days, the individual release kinetics were different. Recovery of specific activity after encapsulation was 40%, 60%, and 90%-that of pre-encapsulation levels for IL-2, GM-CSF and IL-12, respectively. Upon storage, the IL-12 microspheres rapidly lost activity, whereas IL-2 and GM-CSF microspheres remained stable for at least 9 weeks. These studies demonstrate that biochemical properties of microsphere formulations vary depending on the cytokine, and rigorous characterization of formulations is a prerequisite to in vivo testing.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Interleukin-12/chemistry , Interleukin-2/chemistry , Lactic Acid/chemistry , Polymers/chemistry , Adjuvants, Immunologic , Animals , Cells, Cultured , Delayed-Action Preparations , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Humans , Interleukin-12/pharmacokinetics , Interleukin-2/pharmacokinetics , Mice , Microspheres , Pharmaceutical Preparations , Polyesters , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics
11.
Cancer Res ; 63(12): 3202-10, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810649

ABSTRACT

The potent antitumor activity of certain cytokines is often achieved at the expense of unacceptable toxicity. One avenue to improve the therapeutic index of cytokines in cancer therapy consists of fusing them to monoclonal antibodies capable of a selective localization at the tumor site. We have constructed fusion proteins of interleukin-12 (IL-12) and tumor necrosis factor (TNF-alpha) with L19, an antibody fragment specific to the extradomain B of fibronectin which has been shown to target tumors in animal models and in patients with cancer. These fusions display a potent antitumor activity in several immunocompetent murine models of cancer but do not lead to complete remissions of established aggressive tumors. In this article, we have evaluated the tumor-targeting properties and the anticancer activities of combinations of the two antibody-cytokine fusion proteins, as well as of a triple fusion protein between IL-12, L19, and TNF-alpha. Although all fusion proteins were active in vitro, the triple fusion protein failed to localize to tumors in vivo and to show significant therapeutic effects. By contrast, the combination of IL-12-L19 and L19-TNF-alpha displayed potent synergistic anticancer activity and led to the eradication of F9 teratocarcinomas grafted in immunocompetent mice. When cured mice were rechallenged with tumor cells, a delayed onset of tumor growth was observed, indicating the induction of a partial antitumor vaccination effect. Potent anticancer effects were achieved at doses of IL-12-L19 and L19-TNF-alpha (2 micro g + 2 micro g/mouse), which were at least 5-fold lower than the maximal-tolerated dose. The combined administration of the two fusion proteins showed only a modest increase in toxicity, compared with treatments performed with the individual fusion proteins. These results show that the targeted delivery of cytokines to the tumor environment strongly potentiates their antitumor activity and that the combination treatment with IL-12-L19 and L19-TNF-alpha appears to be synergistic in vivo.


Subject(s)
Immunoconjugates/therapeutic use , Immunoglobulin Variable Region/therapeutic use , Immunotherapy , Interleukin-12/therapeutic use , Teratocarcinoma/therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Drug Delivery Systems , Drug Screening Assays, Antitumor , Drug Synergism , Female , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Immunoglobulin Variable Region/administration & dosage , Immunoglobulin Variable Region/toxicity , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Interleukin-12/toxicity , Mice , Neoplasm Transplantation , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/toxicity , T-Lymphocytes/metabolism , Teratocarcinoma/pathology , Tissue Distribution , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/toxicity , Vaccination
12.
J Control Release ; 87(1-3): 177-86, 2003 Feb 21.
Article in English | MEDLINE | ID: mdl-12618034

ABSTRACT

The recruitment of the body's own immune system is amongst the most potent defenses known against cancer. Recent attempts to harness this response have enlisted the use of the immune modulating cytokine, interleukin-12 (IL-12). The objective of this work is to investigate the organ distribution and anti-tumor response in vivo after intratumoral administration of IL-12 expression plasmid complexed with water soluble lipopolymer (WSLP). Formulations of WSLP/p2CMVmIL-12 at N/P mol ratio of 20:1 were prepared in the presence of 5% (w/v) glucose. Organ distribution data following intratumoral injection of CT-26 subcutaneous tumor-bearing BALB/c mice demonstrated enhanced retention of WSLP/p2CMVmIL-12 complexes within the tumor and limited accumulation in other organs for up to 96 h. Tumor-bearing BALB/c mice received either single or repeated intratumoral injections at 4- or 8-day intervals to examine the efficacy of single versus repeated injections on tumor regression and survival. Significant tumor growth inhibition during 4- and 8-day injection trials was observed with maximal survival in mice receiving 4-day injections of WSLP/p2CMVmIL-12 complexes. In conclusion, the water-soluble non-toxic lipopolymer complexed with p2CMVIL-12 showed enhanced transgene expression in vivo, inhibits the rate of tumor growth, and significantly increases survival.


Subject(s)
Drug Delivery Systems/methods , Interleukin-12/administration & dosage , Lipids/administration & dosage , Polymers/administration & dosage , Xenograft Model Antitumor Assays/methods , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cytomegalovirus/genetics , Female , Injections, Intralesional , Interleukin-12/genetics , Interleukin-12/pharmacokinetics , Lipids/pharmacokinetics , Mice , Mice, Inbred BALB C , Plasmids/administration & dosage , Polymers/pharmacokinetics , Solubility/drug effects , Tumor Cells, Cultured , Water/chemistry
13.
Clin Cancer Res ; 8(12): 3686-95, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473577

ABSTRACT

PURPOSE: The purpose is to determine dose-limiting toxicity, pharmacokinetics,pharmacodynamics, and immunobiology after i.p. injections of recombinant human IL-12 (rhIL-12). EXPERIMENTAL DESIGN: rhIL-12 was administered to 29 previously treated patients with peritoneal carcinomatosis from Müllerian carcinomas, gastrointestinal tract carcinomas and peritoneal mesothelioma in a Phase I trial. rhIL-12 doses were increased from 3 to 600 ng/kg. Three or more patients at each level received weekly i.p. injections of rhIL-12. RESULTS: Dose-limiting toxicity (elevated transaminase) occurred in 2 of 4 patients at the 600 ng/kg dose. More frequent toxicities included fever, fatigue, abdominal pain, nausea, and catheter-related infections. Ten patients received 300 ng/kg with acceptable frequency and severity of side effects. Two patients (one with ovarian cancer and one with mesothelioma) had no remaining disease at laparoscopy. Eight patients had stable disease and 19 progressive disease. At 300 ng/kg i.p., IL-12 was cleared from peritoneal fluid in a biphasic manner with a terminal-phase half-life of 18.7 h; peritoneal fluid levels of IL-12 5 min after i.p. injection were 100-200 pg/ml, and serum levels reached approximately 10 pg/ml between 24 and 36 h. IL-1-alpha, IL-2, IL-10, tumor necrosis factor alpha, and IFN-gamma were determined in serum and peritoneal fluid. IFN-gamma, IL-10, and tumor necrosis factor alpha were detected most frequently. Immunobiological effects included peritoneal tumor cell apoptosis, decreased tumor cell expression of basic fibroblast growth factor and vascular endothelial growth factor, elevated IFN-gamma and IFN-inducible protein 10 transcripts in peritoneal exudate cells, and increased proportions of peritoneal CD3(+) relative to CD14(+) cells. CONCLUSIONS: rhIL-12 at 300 ng/kg by weekly i.p. injection is biologically active and adequately tolerated for Phase II studies.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Interleukin-12/therapeutic use , Mesothelioma/drug therapy , Mullerian Ducts/pathology , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Apoptosis/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Gastrointestinal Neoplasms/pathology , Humans , Injections, Intraperitoneal , Intercellular Signaling Peptides and Proteins/metabolism , Interferon-gamma/metabolism , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Lymphokines/metabolism , Male , Mesothelioma/pathology , Middle Aged , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Ploidies , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Tissue Distribution , Transaminases/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Clin Cancer Res ; 8(11): 3383-93, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429625

ABSTRACT

PURPOSE: To determine the safety, maximum tolerated dose,and biological effects of recombinant human IL-12 after autologous stem cell transplantation for cancer. EXPERIMENTAL DESIGN: Twelve patients with hematological malignancies (8 non-Hodgkin's lymphoma, 2 Hodgkin's disease, and 2 plasma cell myeloma) began interleukin (IL)-12 therapy at a median of 66 days after transplantation. Recombinant human IL-12 was given by bolus i.v. injection in doses of 30, 100, or 250 ng/kg once as an inpatient and then, after a 2-week hiatus, once daily for 5 consecutive days every 3 weeks on an outpatient basis. RESULTS: Common side effects included fever, chills, fatigue, nausea or vomiting, and asymptomatic elevation in serum liver function tests. Transient neutropenia and thrombocytopenia were also common, but no patient required platelet transfusion or had a neutropenic fever. Dose-limiting toxicities (diarrhea and elevated liver function tests) occurred in 2 of 3 patients treated in the 250 ng/kg cohort. Biological effects, including increases in serum IFN-gamma levels and transient lymphopenia involving CD4 T cells, CD8 T cells, B cells, and NK cells, were seen at all three dose levels. CONCLUSIONS: Biologically active doses of IL-12 can be given safely to patients after autologous stem cell transplantation for high-risk hematological malignancies. Further studies are indicated to assess the efficacy of IL-12 in this setting.


Subject(s)
Hematologic Neoplasms/therapy , Immunotherapy/methods , Interleukin-12/therapeutic use , Stem Cell Transplantation , Adult , Aged , CD4-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Drug , Female , Hodgkin Disease/therapy , Humans , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-12/pharmacokinetics , Kinetics , Lymphoma, Non-Hodgkin/therapy , Male , Maximum Tolerated Dose , Middle Aged , Plasmacytoma/therapy , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Time Factors , Transplantation, Autologous
15.
J Pharm Pharmacol ; 54(2): 241-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11848288

ABSTRACT

We have investigated the potential toxicity of hlL-12 DNA plasmid formulated with 5% polyvinylpyrrolidone (PVP) administered twice weekly via subcutaneous injections to Cynomolgus monkeys for four weeks, and have evaluated recovery from any effects of the test article over a four-week treatment-free period. Doses of the formulated hIL-12 plasmid were selected based on anti-tumour efficacy studies previously conducted in mice. The duration of the study and the frequency of dosing were designed to support clinical trials. No clinical signs indicative of an adverse effect of administration of formulated hIL-12 plasmid were observed. There were no apparent effects of the formulated hIL-12 plasmid on body weights or on serum chemistry, haematology, coagulation or urinalysis parameters. No treatment-elated ocular abnormalities were evident. In addition, examination of the electrocardiograms from all monkeys at the pre-study, week-4, and week-8 time points did not reveal any treatment-related effects. No treatment-related gross lesions were noted at days 28 or 57. Slight histopathological changes associated with high doses of PVP vehicle were observed at both time points. These results suggested that the administration of formulated hIL-12 plasmid at a dose level up to 18 mg kg(-1) dose twice per week for four weeks to experimentally naïve Cynomolgus monkeys did not result in significant toxicity. These results support further testing of this gene therapy in clinical trials.


Subject(s)
Interleukin-12/toxicity , Povidone/toxicity , Animals , Body Weight/drug effects , DNA/analysis , Electrocardiography/drug effects , Eye/drug effects , Female , Genetic Vectors , Humans , Injections, Subcutaneous , Interleukin-12/chemistry , Interleukin-12/pharmacokinetics , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Plasmids , Povidone/chemistry , Povidone/pharmacokinetics , Skin/chemistry , Toxicity Tests
16.
Hum Gene Ther ; 13(2): 177-85, 2002 Jan 20.
Article in English | MEDLINE | ID: mdl-11812275

ABSTRACT

Interleukin 12 (IL-12) is one of the most effective and promising cytokines for cancer therapy. Its therapeutic effects have been demonstrated in a variety of tumors in animal models when it is administrated locally or systemically. We describe here a systemic delivery of naked murine IL-12 (mIL-12) gene in vivo. Dose-dependent systemic production of mIL-12, with a serum level up to approximately 20 microg/ml, was observed 24 hr after systemic gene delivery. The apparent half-life in the circulation was about 5 hr. The result of a bioactivity assay (in vitro interferon gamma [IFN-gamma] release) indicated that the gene product in mice was as active as the purified recombinant murine IL-12 protein (rmIL-12). The circulating mIL-12 activated natural killer cells and stimulated IFN-gamma production in vivo. A single administration of mIL-12 gene resulted in prominent regression of established subcutaneous tumor in a human papillomavirus (HPV) DNA-positive tumor model (TC-1) in C57BL/6J mice. The antitumor effect of the single gene dose was comparable to repeated intraperitoneal administration of rmIL-12 (0.5 microg/day for consecutive 5 days). This systemic gene delivery is simple, economical, and highly efficient for the production of large amounts of cytokine in vivo. With this gene delivery method, we have demonstrated the therapeutic potential of IL-12 for the treatment of HPV DNA-positive tumor and the usefulness of the systemic gene delivery for assessing the therapeutic effect of a candidate gene.


Subject(s)
DNA/administration & dosage , Genetic Therapy , Interleukin-12/genetics , Papillomavirus Infections/therapy , Tumor Virus Infections/therapy , Animals , DNA/therapeutic use , Gene Dosage , Gene Expression , Humans , Interferon-gamma/metabolism , Interleukin-12/pharmacokinetics , Interleukin-12/therapeutic use , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Papillomaviridae , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured
17.
Antiviral Res ; 52(2): 181-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11672828

ABSTRACT

Interleukin-12 plays a central role in mounting an effective cellular immune response directed towards elimination of intracellular pathogens. In two open label, multicenter, dose-escalation phase I/II studies tolerability, pharmacokinetics, pharmacodynamics, and efficacy of subcutaneously administered recombinant human interleukin-12 (rHuIL-12) was assessed in the treatment of chronic hepatitis B and C. Forty-six patients with chronic hepatitis B and 60 patients with chronic hepatitis C were treated for 12 and 10 consecutive weeks, respectively. rHuIL-12 was generally well tolerated, but was associated with temporary decreases in neutrophils and lymphocyte counts, and with elevations in serum transaminases and bilirubin. Serum IL-12 levels observed were higher at 0.5 microg/kg compared with 0.25 microg/kg doses, suggesting a dose-related increase in systemic exposure of IL-12. Measurable levels of interferon-gamma were also observed at the highest dose of 0.5 microg/kg. At the end of treatment HBV DNA clearance was greater in patients treated with 0.50 microg/kg (25%) or with 0.25 microg/kg (13%) compared with those given 0.03 microg/kg. In patients with chronic hepatitis C, HCV RNA remained detectable in all patients. A more than 50% decrease in pretreatment HCV RNA levels was observed in 3/16 patients (0.03 microg/kg), in 3/14 (0.10 microg/kg), in 6/15 (0.25 microg/kg), and in 8/15 patients of the 0.5 microg/kg dose group. In conclusion, antiviral activity of rHuIL-12 in patients with chronic hepatitis B or C does not appear to be advantageous in comparison to other currently available treatments.


Subject(s)
Hepatitis B, Chronic/drug therapy , Hepatitis C, Chronic/drug therapy , Interleukin-12/therapeutic use , Adolescent , Adult , Female , Hepatitis B, Chronic/blood , Hepatitis C, Chronic/blood , Humans , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
18.
J Interferon Cytokine Res ; 21(4): 257-63, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11359657

ABSTRACT

Recombinant human interleukin-12 (rHuIL-12) is a pleiotropic cytokine with anticancer activity against renal cell carcinoma (RCC) in preclinical models and in a phase I trial. A randomized phase II study of rHuIL-12 compared with interferon-alpha (IFN-alpha) evaluated clinical response for patients with previously untreated, advanced RCC. Patients were randomly assigned 2:1 to receive either rHuIL-12 or IFN-alpha2a. rHuIL-12 was administered by subcutaneous (s.c.) injection on days 1, 8, and 15 of each 28-day cycle. The dose of IL-12 was escalated during cycle 1 to a maintenance dose of 1.25 microg/kg. IFN was administered at 9 million units by s.c. injection three times per week. Serum concentrations of IL-12, IFN-gamma, IL-10, and neopterin were obtained in 10 patients treated with rHuIL-12 after the first full dose of 1.25 microg/kg given on day 15 (dose 3) of cycle 1 and again after multiple doses on day 15 (dose 6) of cycle 2. Thirty patients were treated with rHuIL-12, and 16 patients were treated with IFN-alpha. Two (7%) of 30 patients treated with rHuIL-12 achieved a partial response, and the trial was closed to accrual based on the low response proportion. IL-12 was absorbed rapidly after s.c. drug administration, with the peak serum concentration appearing at approximately 12 h in both cycles. Serum IL-12 concentrations remained stable on multiple dosing. Levels of IFN-gamma, IL-10, and neopterin increased with rHuIL-12 and were maintained in cycle 2. rHuIL-12 is a novel cytokine with unique pharmacologic and pharmacodynamic features under study for the treatment of malignancy and other medical conditions. The low response proportion associated with rHuIL-12 single-agent therapy against metastatic RCC was disappointing, given the preclinical data. Further study of rHuIL-12 for other medical conditions is underway. For RCC, the study of new cytokines is of the highest priority.


Subject(s)
Carcinoma, Renal Cell/therapy , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Interleukin-12/administration & dosage , Interleukin-12/therapeutic use , Kidney Neoplasms/therapy , Recombinant Proteins/administration & dosage , Adult , Aged , Ascites/etiology , Carcinoma, Renal Cell/immunology , Female , Humans , Injections, Subcutaneous , Interferon alpha-2 , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Kidney Neoplasms/immunology , Male , Middle Aged , Neutropenia/etiology , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Stomatitis/etiology
19.
Clin Cancer Res ; 6(5): 1678-92, 2000 May.
Article in English | MEDLINE | ID: mdl-10815886

ABSTRACT

The aim of this study was to examine the tolerability, antitumor activity, and biological effects of a new schedule of i.v. recombinant human interleukin 12 (rhIL-12). Twenty-eight patients were enrolled in a Phase I trial in which rhIL-12 was administered twice weekly as an i.v. bolus for 6 weeks. Stable or responding patients were eligible to receive additional 6-week cycles until there was no evidence of disease or until tumor progression. Patient cohorts were treated with escalating doses of rhIL-12 (30-700 ng/kg). The maximum tolerated dose (MTD) was 500 ng/kg, with dose-limiting toxicities consisting of elevated hepatic transaminases and cytopenias. At the MTD (n = 14), there was one partial response occurring after 6 cycles of rhIL-12 in a patient with renal cell cancer. Two additional renal cell cancer patients treated at the MTD had prolonged disease stabilization, with one of these exhibiting tumor regression after 8 cycles of rhIL-12. IFN-gamma, IL-15, and IL-18 were induced in patients treated with rhIL-12. Whereas IFN-gamma and IL-15 induction were attenuated midway through the first cycle in patients with disease progression, those patients with tumor regression or prolonged disease stabilization were able to maintain IFN-gamma, IL-15, and IL-18 induction. The down-modulation of IFN-gamma induction during rhIL-12 treatment did not relate to IL-10 production or alterations in rhIL-12 bioavailability but was associated with an acquired defect in lymphocyte IFN-gamma production in response to IL-12, IL-2, or IL-15. This defect could be partially overcome in vitro through combined stimulation with IL-12 plus IL-2. These findings show that the chronic administration of twice-weekly i.v. rhIL-12 is well-tolerated, stimulates the production of IL-12 costimulatory cytokines and IFN-gamma, and can induce delayed tumor regression. Strategies aimed at maintaining IFN-gamma induction, such as the addition of IL-2, may further augment the response rate to this schedule of rhIL-12.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Interleukin-12/therapeutic use , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Adult , Aged , Arthralgia/chemically induced , Carcinoma, Renal Cell/pathology , Dose-Response Relationship, Drug , Female , Fever/chemically induced , Hematologic Diseases/chemically induced , Humans , Hypotension/chemically induced , Injections, Intravenous , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Interleukin-15/metabolism , Interleukin-18/metabolism , Kidney Neoplasms/pathology , Male , Melanoma/pathology , Middle Aged , Mouth Mucosa , Neoplasm Metastasis , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Skin/drug effects , Skin/pathology , Stomatitis/chemically induced , Time Factors
20.
J Hepatol ; 32(2): 317-24, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10707873

ABSTRACT

BACKGROUND/AIMS: Interleukin-12 (IL-12) may be active against hepatitis B virus (HBV). The objective of the study was to assess the tolerability, activity, pharmacokinetics, and pharmacodynamics of three dose levels (0.03 microg/kg b.w., n=15; 0.25 microg/kg b.w., n=15; 0.50 microg/kg b.w., n=16) of recombinant human (rHu) IL-12 given s.c. once a week for 12 consecutive weeks. METHODS: Forty-six patients with chronic hepatitis B, HBV DNA positivity and aminotransferase elevation were included in a multicenter prospective randomized phase I/II study. RESULTS: Compared with the baseline, HBV DNA levels had decreased significantly at the end of rHuIL-12 treatment and after the 12-week follow-up period (p<0.001). The response to rHuIL-12 treatment was dose-dependent: at the end of the study HBV DNA clearance was greater in patients treated with 0.50 microg/kg b.w. (25%) or with 0.25 microg/kg b.w. (13%) compared with those given 0.03 microg/kg b.w. (7%). Moreover, HBeAg became undetectable at the end of follow-up in five of the patients given the 0.25microg/kg (2/15) or the 0.50 microg/kg (3/16) dose. The drug pharmacology showed that IL-12 had an estimated half-life of 30 h with levels remaining detectable for more than 48 h after rHuIL-12 administration. The serum levels of IL-12, interferon-gamma, IL-10, neopterin and beta2-microglobulin as well as the area under the curve (AUC) were rHuIL-12 dose-related. Side effects were observed more frequently with higher doses, including moderate decreases in lymphocyte and neutrophil counts; three patients withdrew prematurely from treatment. The local reaction observed at the injection site was unrelated to the drug dose. Only one patient showed detectable antibody levels to rHuIL-12 without clinical impact. CONCLUSIONS: Treatment with rHuIL-12 at the doses investigated is safe and tolerable, and appears to be active against HBV in patients with chronic hepatitis B.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hepatitis B, Chronic/drug therapy , Interleukin-12/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Female , Hepatitis B, Chronic/blood , Humans , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Interleukin-12/therapeutic use , Male , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Treatment Outcome , beta 2-Microglobulin/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...