Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
J Appl Toxicol ; 44(2): 301-312, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37770382

RESUMO

Interleukin-12 (IL-12) is known to exert antitumor immune effects by promoting the activation and proliferation of T cells and NK cells within the immune system. However, clinical trials have observed systemic toxicity associated with the administration of IL-12. This has shelved development plans for its use as a cancer therapeutic drug. Therefore, it is critical that we perform a systematic evaluation of the toxicity and safety of repeated IL-12 administration. In this study, we conducted a comprehensive evaluation of the toxicity and safety of repeated rhIL-12 (recombinant human interleukin-12) administration in rhesus monkeys by assessing its effects on the immune system, organ function, and vital signs. Rhesus monkeys were subcutaneously injected with 0.5, 2.5, and 12.5 µg/kg of rhIL-12 for up to for 14 consecutive weeks. The low dose exhibited no signs of toxicity, whereas animals receiving higher doses displayed symptoms such as loose stools, reduced activity, anemia, and elevated liver function indicators (AST and TBIL). Following three administrations of 12.5 µg/kg, high dosing was adjusted to 7.5 µg/kg due to manifestations of symptoms like loose stools, decreased activity, and huddling in the cage. Furthermore, rhesus monkeys exhibited marked immunogenic responses to recombinant human interleukin-12 (rhIL-12). However, based on overall study findings, the No Observed Adverse Effect Level (NOAEL) for the subcutaneous injection of rhIL-12, when repeatedly administered for 3 months in rhesus monkeys, was considered to be 0.5 µg/kg. The Highest Non-Severely Toxic Dose (HNSTD) was considered to be 7.5 µg/kg.


Assuntos
Antineoplásicos , Interleucina-12 , Animais , Humanos , Macaca mulatta , Proteínas Recombinantes/toxicidade , Interleucina-12/toxicidade , Células Matadoras Naturais
2.
Environ Toxicol ; 39(5): 3188-3197, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38356236

RESUMO

Yin chai hu (Radix Stellariae) is a root medicine that is frequently used in Chinese traditional medicine to treat fever and malnutrition. In modern medicine, it has been discovered to have anti-inflammatory, anti-allergic, and anticancer properties. In a previous study, we were able to extract lipids from Stellariae Radix using supercritical CO2 extraction (SRE), and these sterol lipids accounted for up to 88.29% of the extract. However, the impact of SRE on the development of atopic dermatitis (AD) has not yet been investigated. This study investigates the inhibitory effects of SRE on AD development using a 2,4-dinitrochlorobenzene (DNCB)-induced AD mouse model. Treatment with SRE significantly reduced the dermatitis score and histopathological changes compared with the DNCB group. The study found that treatment with SRE resulted in a decrease of pro-inflammatory cytokines TNF-α, CXC-10, IL-12, and IL-1ß in skin lesions. Additionally, immunohistochemical analysis revealed that SRE effectively suppressed M1 macrophage infiltration into the AD lesion. Furthermore, the anti-inflammatory effect of SRE was evaluated in LPS + INF-γ induced bone marrow-derived macrophages (BMDMs) M1 polarization, SRE inhibited the production of TNF-α, CXC-10, IL-12, and IL-1ß and decreased the expression of NLRP3. Additionally, SRE was found to increase p-AMPKT172, but had no effect on total AMPK expression, after administration of the AMPK inhibitor Compound C, the inhibitory effect of SRE on M1 macrophages was partially reversed. The results indicate that SRE has an inhibitory effect on AD, making it a potential therapeutic agent for this atopic disorder.


Assuntos
Dermatite Atópica , Animais , Camundongos , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/metabolismo , Dinitroclorobenzeno/toxicidade , Dinitroclorobenzeno/uso terapêutico , Proteínas Quinases Ativadas por AMP , Dióxido de Carbono/toxicidade , Dióxido de Carbono/uso terapêutico , Fator de Necrose Tumoral alfa , Citocinas/metabolismo , Macrófagos/metabolismo , Anti-Inflamatórios/uso terapêutico , Interleucina-12/toxicidade , Interleucina-12/uso terapêutico , Lipídeos , Camundongos Endogâmicos BALB C , Pele
3.
Anticancer Drugs ; 25(3): 282-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24300915

RESUMO

The aim of this study was to examine the hypothesis that a combination of proteasome inhibition by bortezomib and immune therapy with interleukin-12 (IL-12) can produce enhanced antitumor efficacy relative to the effects of either of these agents alone. A mouse xenograft model of myeloma was developed. The mice were randomly divided into saline control (NS), IL-12 (0.4 µg/animal; intraperitoneal), bortezomib (0.75 mg/kg; intravenous), and bortezomib+IL-12 groups. Effects of treatments on tumor growth were assessed by before and after treatment comparisons and group comparisons. The effects of various treatments on the number of peripheral blood lymphocytes and natural killer (NK) cells were assessed by complete blood count and flow cytometry analysis. The cell-killing function of NK cells in splenocytes was evaluated using the lactate dehydrogenase release assay. IL-12 treatment alone produced a mild decrease in tumor volume compared with control (P>0.05). Bortezomib alone resulted in substantial inhibition of tumor growth at varying time points, reaching ~65 and ~60% reduction in tumor volume after 15 and 21 days of therapy, respectively. At the same time points, the combination therapy produced ~75 and ~84% decreases in tumor growth, respectively, which were significantly greater than the reduction produced by bortezomib monotherapy. Tumors resumed growth upon termination of bortezomib treatment at 2 weeks, although the tumor volume was still significantly smaller than that in the time-matched NS and IL-12 animals. This rebound of tumor growth was completely prevented with the combination therapy, and tumor volume continued to decrease throughout the time course. The percentage and total number of NK cells were significantly decreased after bortezomib monotherapy and combination therapy; however, they remained unaltered after IL-12 treatment compared with no treatment. Further, combination therapy significantly restored the bortezomib-induced functional impairment of the cell-killing capability of NK cells, relative to bortezomib alone. We conclude that the bortezomib-IL-12 combination therapy offers superior antitumor efficacy over monotherapy with either bortezomib or IL-12 in a mouse model of myeloma. Restoration of bortezomib-induced functional impairment of NK cells by IL-12 may be a mechanism for the synergetic effects of the two agents. Therefore, a combination of the two agents may represent a more rational therapeutic approach for myeloma.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Interleucina-12/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Pirazinas/farmacologia , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Apoptose , Ácidos Borônicos/uso terapêutico , Ácidos Borônicos/toxicidade , Bortezomib , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Xenoenxertos , Humanos , Imunoterapia , Interleucina-12/uso terapêutico , Interleucina-12/toxicidade , Masculino , Camundongos , Camundongos SCID , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Pirazinas/uso terapêutico , Pirazinas/toxicidade
4.
Adv Mater ; 36(19): e2309927, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38387609

RESUMO

Cytokines are powerful in cancer immunotherapy, however, their therapeutic potential is limited by the severe systemic toxicity. Here a potent strategy to reduce the toxicity of systemic cytokine therapy by delivering its denatured form using a finely designed nanochaperone, is described. It is demonstrated that even if the denatured protein cargos are occasionally released under normal physiological conditions they are still misfolded, while can effectively refold into native states and release to function in tumor microenvironment. Consequently, the systemic toxicity of cytokines is nearly completely overcome. Moreover, an immunogenic cell death (ICD)-inducing chemotherapeutic is further loaded and delivered to tumor using this nanochaperone to trigger the release of tumor-associated antigens (TAAs) that are subsequently captured in situ by nanochaperone and then reflows into lymph nodes (LNs) to promote antigen cross-presentation. This optimized personalized nanochaperone-vaccine demonstrates unprecedented suppressive effects against large, advanced tumors, and in combination with immune checkpoint blockade (ICB) therapy results in a significant abscopal effect and inhibition of postoperative tumor recurrence and metastasis. Hence, this approach provides a simple and universal delivery strategy to reduce the systemic toxicities of cytokines, as well as provides a robust personalized cancer vaccination platform, which may find wide applications in cancer immunotherapy.


Assuntos
Antígenos de Neoplasias , Imunoterapia , Interleucina-12 , Nanoestruturas , Animais , Humanos , Camundongos , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/química , Linhagem Celular Tumoral , Morte Celular Imunogênica/efeitos dos fármacos , Imunoterapia/métodos , Interleucina-12/química , Interleucina-12/metabolismo , Interleucina-12/toxicidade , Nanopartículas/química , Nanoestruturas/química , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Dobramento de Proteína , Microambiente Tumoral/efeitos dos fármacos
5.
J Immunol ; 183(1): 740-8, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535628

RESUMO

IL-12 is an excellent candidate for the treatment of cancer due to its ability to drive strong antitumor responses. Recombinant IL-12 protein is currently used in cancer patients; however, systemic expression of rIL-12 presents disadvantages including cost and dose limitation due to its toxicity. In this study, we used hydrodynamic shear of cDNA as a tool to achieve systemic expression of IL-12. We found that sustained but toxic levels of serum IL-12 could be generated in 6- to 7-wk-old B6 mice after a single injection of the cDNA. Unexpectedly, we observed that when IL-12 cDNA is coinjected with IL-18 cDNA, IL-12 antitumor activity was maintained, but there was a significant attenuation of IL-12 toxicity, as evidenced by a greater survival index and a diminution of liver enzymes (ALT and AST). Interestingly, after IL-12 plus IL-18 cDNA administration, more rapid and higher IL-10 levels were observed than after IL-12 cDNA treatment alone. To understand the mechanism of protection, we coinjected IL-12 plus IL-10 cDNAs and observed an increase in survival that correlated with diminished serum levels of the inflammatory cytokines TNF-alpha and IFN-gamma. Confirming the protective role of early IL-10 expression, we observed a significant decrease in survival in IL-10 knockout mice or IL-10R-blocked B6 mice after IL-12 plus IL-18 treatment. Thus, our data demonstrate that the high and early IL-10 expression induced after IL-12 plus IL-18 cDNA treatment is critical to rapidly attenuate IL-12 toxicity without affecting its antitumor capacity. These data could highly contribute to the design of more efficient/less toxic protocols for the treatment of cancer.


Assuntos
Antineoplásicos/metabolismo , Interleucina-10/biossíntese , Interleucina-10/uso terapêutico , Interleucina-12/biossíntese , Interleucina-12/toxicidade , Interleucina-18/biossíntese , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Animais , Antineoplásicos/sangue , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , DNA Complementar/administração & dosagem , DNA Complementar/biossíntese , Quimioterapia Combinada , Interferon gama/antagonistas & inibidores , Interferon gama/sangue , Interferon gama/deficiência , Interleucina-10/deficiência , Interleucina-12/sangue , Interleucina-12/uso terapêutico , Interleucina-18/fisiologia , Interleucina-18/uso terapêutico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/mortalidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue
6.
J Exp Med ; 181(3): 901-14, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7869050

RESUMO

Interleukin 12 (IL-12) doses in excess of 100 ng/d have been shown to induce profound immunotoxicities in mice infected with lymphocytic choriomeningitis virus (LCMV). These immunotoxicities are characterized by almost complete inhibition of virus-induced CD8+ T cell expansion and CTL activation, and up to 2 log increases in viral replication. They are accompanied by induction of serum tumor necrosis factor (TNF). The studies presented here were undertaken to characterize mechanisms for the IL-12-induced toxicities and to examine expression and function of TNF in this context. Several physiological changes were induced in IL-12-treated uninfected and dramatically elevated in IL-12-treated virus-infected mice. IL-12 induced (a) decreases in body weights, > 10% in uninfected and > 20% in LCMV-infected mice; (b) elevation of circulating glucocorticoid levels to > 10 micrograms/dl in uninfected and > 20 micrograms/dl in infected mice; and (c) decreases in thymic mass, > 30% in uninfected and up to 95% in infected mice. These changes are known to be associated with circulating TNF. Northern blot and in situ hybridization analyses demonstrated that IL-12 induced TNF-alpha expression and that LCMV infection synergized with IL-12 for induction of this factor. Antibodies neutralizing TNF reversed all of the IL-12-induced toxicities in LCMV-infected mice including the immunotoxicities against CD8+ T cells and anti-viral defenses. The TNF-mediated immunotoxicities appeared to result from an induced cellular sensitivity to the factor, as splenic leukocytes and CD8+ T cell subsets isolated from LCMV-infected mice were more sensitive to TNF-mediated cytotoxicity in culture than were equivalent populations prepared from uninfected mice. Experiments with the glucocorticoid type II receptor antagonist, RU486, demonstrated that endogenous glucocorticoids were secondary intermediaries in IL-12-induced thymic atrophy. Studies in IL-2-deficient mice showed that the synergism was dependent upon endogenous IL-2. The results delineate a unique mechanism of TNF-mediated toxicity. In addition, they have significant implications concerning potential detrimental consequences of in vivo TNF induction and of IL-12 administration for protective anti-viral responses.


Assuntos
Glucocorticoides/fisiologia , Interleucina-12/toxicidade , Coriomeningite Linfocítica/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Linfócitos T CD8-Positivos/fisiologia , Corticosterona/sangue , Interleucina-2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mifepristona/farmacologia
7.
Curr Opin Immunol ; 10(3): 259-64, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9638361

RESUMO

IL-18 (interferon-inducing factor) and IL-12 exhibit a marked synergism in interferon-gamma induction in T cells. Investigations into the mechanism of this synergism have revealed that IL-12 upregulates expression of the IL-18 receptor on cells producing interferon-gamma. Although IL-18 does not induce the development of Th1 cells, it is essential for the effective induction and activation of Th1 cells by IL-12. As for natural killer cells, IL-18 seems to activate them independently of IL-12. Although IL-12 and IL-18 activate both innate and acquired immunity, their excessive production by activated macrophages may induce multiple organ disorders including disruption of the immune system.


Assuntos
Citocinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/biossíntese , Interleucina-12/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Células Cultivadas , Citocinas/fisiologia , Citocinas/toxicidade , Sinergismo Farmacológico , Imunoglobulina E/biossíntese , Interferon gama/genética , Interleucina-12/fisiologia , Interleucina-12/toxicidade , Interleucina-18 , Subunidade alfa de Receptor de Interleucina-18 , Células Matadoras Naturais/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Imunológicos , RNA Mensageiro/biossíntese , Receptores de Interleucina/biossíntese , Receptores de Interleucina/genética , Receptores de Interleucina-18 , Transdução de Sinais , Linfócitos T/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Regulação para Cima/efeitos dos fármacos
8.
J Transl Med ; 5: 66, 2007 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18076766

RESUMO

BACKGROUND: Pharmacokinetic advantages of intraperitoneal (IP) rhIL-12, tumor response to IP delivery of other cytokines as well as its potential anti-angiogenic effect provided the rationale for further evaluation of IPrhIL-12 in patients with persistent ovarian or peritoneal carcinoma. METHODS: A phase 2 multi-institutional trial (NCI Study #2251) of IP rIL-12 (300 nanogram/Kg weekly) was conducted in patients with ovarian carcinoma or primary peritoneal carcinoma. Patients treated with primary therapy for ovarian cancer who had no extraabdominal/parenchymal disease or bulky peritoneal disease were eligible. Four to 8 weeks from last chemotherapy, eligible patients underwent a laparotomy/laparoscopy. Patients with residual disease < or = 1 cm were registered for the treatment phase 2-5 weeks post surgery. The effect of IP rIL-12 on the expression of TNFalpha , INFalpha , IL-10, IP-10, IL-8, FGF, VEGF was also studied. RESULTS: Thirty-four patients were registered for the first screening phase of the study. Median age was 56.6 years (range: 31-71); 12 completed the second phase and were evaluable for response/toxicity. Performance scores of IL-12 treated patients were 0 (11 pts) and 1 (1 pt). There were no treatment related deaths, peritonitis or significant catheter related complications. Toxicities included grade 4 neutropenia (1), grade 3 fatigue (4), headache (2), myalgia (2), non-neutropenic fever (1), drug fever (1), back pain (1), and dizziness (1). The best response observed was SD. Two patients had SD and 9 had PD, and 1 was evaluable for toxicity only. Peritoneal fluid cytokine measurements demonstrated a > or = 3 fold relative increase post-rhIL-12: IFN-gamma, 5/5 pts; TNF-alpha , 1/5; IL-10, 4/5; IL-8, 5/5; and VEGF, 3/5. IP10 levels were increased in 5/5 patients. Cytokine response profiles suggest either NK or T-cell mediated effects of IP rhIL-12. Cytokine/chemokine results also suggest a pleiotropic response since proteins with potential for either anti-tumor (IFN-gamma , IP-10) or pro-tumor growth effects (VEGF, IL-8) were detected. CONCLUSION: IP IL-12 can safely be administered at this dose and schedule to patients after first line chemotherapy for ovarian/peritoneal carcinoma. The maximum response was stable disease. Future IP therapies with rhIL-12 will require better understanding and control of pleiotropic effects of IL-12.


Assuntos
Interleucina-12/toxicidade , Interleucina-12/uso terapêutico , Neoplasia Residual/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Adulto , Idoso , Líquido Ascítico/química , Ensaios Clínicos Fase I como Assunto , Citocinas/análise , Feminino , Humanos , Pessoa de Meia-Idade , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Segurança
9.
Clin Cancer Res ; 12(20 Pt 1): 6056-63, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17062681

RESUMO

PURPOSE: Rituximab is a chimeric antibody that induces B-cell apoptosis and recruits immune effector cells to mediate cell lysis. Interleukin-12 (IL-12) facilitates cytolytic responses by T cells and natural killer cells. This phase II study was done to determine the efficacy and toxicity of IL-12 in combination with rituximab in patients with B-cell non-Hodgkin's lymphoma (NHL). EXPERIMENTAL DESIGN: Fifty-eight patients with histologically confirmed relapsed B-cell NHL were randomized to receive concurrent treatment with rituximab and IL-12 (arm A) or rituximab with subsequent treatment with IL-12 after documented nonresponse or progression after rituximab (arm B). Treatment consisted of 375 mg/m2 rituximab on days 1, 8, 15, and 22 and 300 ng/kg IL-12 given s.c. twice weekly starting on day 2 for arm A or upon progression for arm B. RESULTS: The overall response rate was 37% (11 of 30) in arm A and 52% (13 of 25) in arm B. All of the responses seen in arm B occurred while patients received rituximab, and no responses occurred during treatment with subsequent IL-12. The median duration of response was 16 months for arm A and 12 months for arm B. Biopsy specimens were serially obtained in a subset of patients and showed that changes in gene expression were different when cells from the peripheral blood were compared with cells from lymph node biopsies. CONCLUSIONS: The concomitant use of IL-12 and rituximab had modest disease activity in patients with B-cell NHL, but the sequential administration of IL-12 after rituximab did not result in additional clinical responses.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Interleucina-12/uso terapêutico , Linfoma de Células B/tratamento farmacológico , Linfoma não Hodgkin/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais Murinos , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Esquema de Medicação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-12/toxicidade , Linfoma de Células B/genética , Linfoma de Células B/imunologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/imunologia , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Seleção de Pacientes , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação , Rituximab , Linfócitos T/imunologia
10.
J Interferon Cytokine Res ; 26(9): 593-608, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16978064

RESUMO

It has become increasingly apparent that the ability to generate an optimal host immune response requires effective cross talk between the innate and adaptive components of the immune system. Pro-inflammatory cytokines, in particular those that can induce a danger signal, often called signal 3, are crucial in this role of initiating and augmenting the presentation of exogenous antigen to T cells by dendritic cells. Interleukin-12 (IL-12) in particular has been defined as a "signal 3" cytokine required for the antigen cross priming. Given this unique interactive function, a significant amount of work has been performed to define possible therapeutic applications for IL-12. Systemic IL-12 administration can clearly act as a potent adjuvant for postvaccination T cell responses in a variety of diseases. As an example, in the cancer setting, systemic IL-12 is capable of suppressing tumor growth, metastasis, and angiogenesis in vivo. IL-12, however, has been associated with significant dose- and schedule-dependent toxicity in early clinical trials, results that have proven to be a major obstacle to its clinical application. Recent research has focused on decreasing the toxicity of IL-12 using different delivery approaches, including virus-based and gene-modified cell-based delivery. Although effective, these approaches also have limitations, including the generation of neutralizing antibodies, in addition to lacking the simplicity and versatility required for universal clinical application. Thus, there is a significant interest in the development of alternative delivery approaches for IL-12 administration that can overcome these issues. Several nonviral delivery approaches for IL-12 protein or gene expression vectors are being defined, including alum, liposomes, and polymer-based delivery. These developing approaches have shown promising adjuvant effects with significantly lessened systemic toxicity. This article discusses the potential capabilities of these nonvirus-based IL-12 delivery systems in different disease settings, including allergy, infection, and cancer.


Assuntos
Adjuvantes Imunológicos , Técnicas de Transferência de Genes , Imunoterapia/métodos , Interleucina-12 , Compostos de Alúmen/metabolismo , Animais , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/terapia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-12/uso terapêutico , Interleucina-12/toxicidade , Lipossomos/metabolismo , Neoplasias/imunologia , Neoplasias/terapia
11.
Cancer Res ; 57(12): 2460-7, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9192826

RESUMO

Use of the cytokine interleukin 12 (IL-12) has been shown to enhance the rejection of a variety of murine tumors, but preclinical and clinical studies have revealed that recombinant IL-12 (rlL-12) can produce severe toxicity. In an effort to improve the tolerance and therapeutic effectiveness of this cytokine, we investigated the influence of giving a single dose of recombinant murine IL-12 (rmIL-12) a week prior to daily cytokine administration (predosing) on its toxic and antitumor effects. These studies were performed in C3H/HeN mice, in which a course of rmIL-12 at standard doses without predosing induced rejection of syngeneic K1735 melanomas in 33%, and in A/J mice, in which treatment induced rejection of syngeneic B7-1+ SCK (SCK.B7-1) mammary carcinomas in 63%. Administration of a predose of rmIL-12 markedly reduced cytokine toxicity in a dose-dependent manner and allowed safe administration of up to 8-fold higher doses of daily rmIL-12 in C3H/HeN mice and 4-fold higher doses of rmIL-12 in A/J mice. Predosing followed by either standard or high daily doses of rmIL-12 did not significantly alter most end points of rmIL-12 treatment of K1735 or SCK.B7-1 tumors (survival, death from tumor, development of protective immunity, and so on), but they appeared to attenuate early control of tumorigenesis by rmIL-12. Evidence for the latter comes from a shortening of the characteristic rmIL-12-induced delay in tumor appearance and in the frequent appearance of tumors that subsequently regress. However, higher doses appear to produce better therapeutic results than standard doses of rmIL-12 after predosing. Predosing severely blunted induction of serum IFN-gamma levels by rmIL-12, which probably accounts for many of the effects of predosing on rmIL-12 toxicity and efficacy. Thus, predosing desensitizes mice to the toxic effects of rIL-12 and allows much higher doses to be given but, despite this, it does not improve and, by some criteria, it attenuates rIL-12 therapeutic outcome. Our results do not support the use of predosing as a way to enhance the effectiveness of rIL-12 in cancer clinical trials.


Assuntos
Antineoplásicos/toxicidade , Interleucina-12/administração & dosagem , Interleucina-12/toxicidade , Proteínas Recombinantes/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Feminino , Interferon gama/sangue , Interleucina-12/sangue , Neoplasias Mamárias Experimentais/tratamento farmacológico , Melanoma/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos , Transplante de Neoplasias , Proteínas Recombinantes/sangue , Proteínas Recombinantes/toxicidade , Células Tumorais Cultivadas
12.
Cancer Res ; 63(12): 3202-10, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12810649

RESUMO

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.


Assuntos
Imunoconjugados/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Imunoterapia , Interleucina-12/uso terapêutico , Teratocarcinoma/terapia , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Imunoconjugados/administração & dosagem , Imunoconjugados/farmacocinética , Imunoconjugados/toxicidade , Região Variável de Imunoglobulina/administração & dosagem , Região Variável de Imunoglobulina/toxicidade , Interferon gama/metabolismo , Interleucina-12/administração & dosagem , Interleucina-12/farmacocinética , Interleucina-12/toxicidade , Camundongos , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes de Fusão/toxicidade , Linfócitos T/metabolismo , Teratocarcinoma/patologia , Distribuição Tecidual , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacocinética , Fator de Necrose Tumoral alfa/toxicidade , Vacinação
13.
Clin Cancer Res ; 10(16): 5432-8, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15328181

RESUMO

PURPOSE: The purpose of this study was to evaluate the clinical activity and toxicity of recombinant human Interleukin (IL)-12 in patients with relapsed and refractory non-Hodgkin's lymphoma (NHL) or Hodgkin's disease (HD). EXPERIMENTAL DESIGN: Forty-two previously treated patients (32 patients with NHL and 10 patients with HD) were enrolled on the study. Patients were treated with either intravenous (n = 11) or subcutaneous (n = 31) administration of IL-12. The patients had received a median of three prior treatment regimens, and 16 patients had undergone prior autologous stem cell transplantation. RESULTS: All patients were assessable for toxicity, and 39 of 42 (93%) patients were assessable for response. Six of 29 (21%) patients with NHL had a partial or complete response, whereas none of the 10 patients with HD responded. Furthermore, 15 patients had stable disease that lasted for up to 54 months. Progression-free survival in patients with indolent NHL, aggressive NHL, and HD was 6, 2, and 2.5 months, respectively. Treatment was well tolerated, and the most common toxicity was flu-like symptoms. Reversible grade 3 hepatic toxicity was observed in three patients requiring dose reduction. IL-12 therapy increased the median number of peripheral blood CD8 T lymphocytes from 423/microl to 576/microl (P = 0.0019). Furthermore, IL-12 therapy decreased serum vascular endothelial growth factor and basic fibroblast growth factor concentrations in 37% of the patients. CONCLUSIONS: The ability of recombinant human IL-12 therapy to increase the number of circulating CD8+ cells and induce clinical remissions in patients with relapsed NHL warrants further investigation of the drug.


Assuntos
Doença de Hodgkin/tratamento farmacológico , Interleucina-12/toxicidade , Interleucina-12/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Interleucina-12/administração & dosagem , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Recidiva , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento
14.
Hum Gene Ther ; 10(3): 407-17, 1999 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-10048393

RESUMO

Intramuscular injection of plasmid DNA encoding both subunits of the cytokine interleukin 12 (IL-12) exhibits strong antimetastatic activity against lung metastases induced by the malignant melanoma cell line B16-F10. The protective effect of IL-12 DNA is long-lasting, since administration of tumor cells 9 days after IL-12 DNA treatment prevented metastasis formation. No effects were observed with empty plasmid controls, DNA encoding the melanoma-associated antigen pmel17/gp100, the granulocyte-macrophage colony-stimulating factor GM-CSF, B7.1, or CpG-containing oligodeoxynucleotides. IL-12 DNA is required during early phases of metastasis formation and is ineffective when administered later. Its efficiency is dose dependent. The cytotoxic T cell response contributes to the antimetastatic effect as evidenced by genetically modified CD8- or perforin knockout mice. Depletion of natural killer (NK) cells by antibodies completely abrogated the effect. In contrast, the IL-12-induced antimetastatic effect was not mediated by interferon gamma (IFN-gamma) or tumor necrosis factor alpha (TNF-alpha) as shown with IFN-gamma receptor and TNF-alpha knockout mice, respectively. Toxic side effects by IL-12 were low. Our results suggest that plasmid DNA encoding IL-12 might have potential value as gene medicine against the initiation of metastasis formation.


Assuntos
Técnicas de Transferência de Genes , Interleucina-12/genética , Interleucina-12/farmacologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Animais , Linfócitos T CD8-Positivos/imunologia , Ensaio de Imunoadsorção Enzimática , Injeções Intramusculares , Interleucina-12/toxicidade , Neoplasias Pulmonares/imunologia , Melanoma/genética , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Tempo , Células Tumorais Cultivadas
15.
J Interferon Cytokine Res ; 23(1): 47-50, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12639298

RESUMO

Early and sustained treatment with interleukin-12 (IL-12) ameliorated disease in a mouse model of infection with the encephalitogenic flavivirus, St. Louis encephalitis virus (SLEV, Japanese encephalitis serogroup). However, this effect was not reproduced in murine infections with either the flavivirus tick-bore encephalitis virus (TBEV) or the alphavirus Venezuelan equine encephalitis virus (VEEV). IL-12 exacerbated TBEV disease when used in conjunction with monoclonal antibody (mAb), suggesting an enhancement of immunopathology, and was without clinical effects in VEEV infection. These data confirm the need to fully understand the pathogenesis of viral infection before cytokine intervention may be employed as a broad-spectrum antiviral therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Encefalite de St. Louis/tratamento farmacológico , Encefalite Transmitida por Carrapatos/tratamento farmacológico , Encefalomielite Equina Venezuelana/tratamento farmacológico , Interleucina-12/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/toxicidade , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Antivirais/toxicidade , Modelos Animais de Doenças , Interleucina-12/administração & dosagem , Interleucina-12/toxicidade , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade
16.
J Interferon Cytokine Res ; 23(3): 155-62, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12716488

RESUMO

Acute fatty degeneration in the liver is caused by various agents, such as aspirin, valproic acid, and ibuprofen, that directly inhibit mitochondrial beta-oxidation of fatty acid and oxidative phosphorylation. Endogenous molecules, such as cytokines and hormones, are also known to mediate microvesicular steatosis in liver failure. In this study, we examined how interleukin-12 (IL-12) and IL-18 cause steatosis in the liver. Administration of these cytokines in combination caused marked hepatosteatosis and weight loss in mice. There were marked increases in levels of interferon-gamma (IFN-gamma), nitrite (NO(2)/NO(3)), and fibrinogen in the circulation in these mice. On the other hand, the ATP concentration and blood flow in the liver were significantly reduced. These changes, except the production of IFN-gamma and NO, were partially inhibited by Z-VAD-fmk, a synthetic tripeptide inhibitor for NO-induced caspases. These results indicate that IL-12 and IL-18 may mediate inflammatory hepatosteatosis through impairment of the microcirculation, which leads to mitochondrial dysfunction in hepatocytes.


Assuntos
Fígado Gorduroso/induzido quimicamente , Interleucina-12/toxicidade , Interleucina-18/toxicidade , Fígado/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Fibrinogênio/metabolismo , Interferon gama/biossíntese , Interferon gama/metabolismo , Fluxometria por Laser-Doppler , Fígado/metabolismo , Fígado/patologia , Testes de Função Hepática , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Mitocôndrias Hepáticas/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Nitritos/sangue , Nitritos/metabolismo , Oxirredução , Fluxo Sanguíneo Regional , Fatores de Tempo
18.
Int Immunopharmacol ; 3(6): 801-9, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12781697

RESUMO

Interleukin-12 (IL-12) is a heterodimeric cytokine that enhances immune responses to bacterial, parasitic, and viral pathogens, and leads to tumor regression in animal models. For this reason, the use of IL-12 as a vaccine adjuvant and as a therapeutic agent for the treatment of cancer is being investigated. Unfortunately, the extreme toxicity of this molecule observed during clinical trials has limited its use. This toxicity correlates with increased IFN-gamma expression, decreased glucose levels, and altered histological responses in the spleen and duodenum. In this study, we show that intranasal (i.n.) delivery of IL-12 is a less toxic route of inoculation compared to the commonly employed subcutaneous route. When delivered i.n., IL-12 induces less systemic IFN-gamma production and fewer pathological tissue changes, yet is efficacious, as indicated by enhanced CD3(+) T cell activation and increased production of Th1-associated immunoglobulins (i.e., serum IgG2a). Thus, IL-12 can be delivered safely and effectively by the i.n. route, a finding which may allow IL-12 to fulfill its clinical potential.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/toxicidade , Interleucina-12/administração & dosagem , Interleucina-12/toxicidade , Administração Intranasal , Animais , Complexo CD3/imunologia , Citocinas/biossíntese , Citocinas/genética , Duodeno/metabolismo , Ensaio de Imunoadsorção Enzimática , Imunoglobulina G/imunologia , Imuno-Histoquímica , Injeções Subcutâneas , Interferon gama/biossíntese , Interferon gama/genética , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Baço/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
19.
Eur Cytokine Netw ; 11(1): 67-74, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10705301

RESUMO

There is increasing evidence that IL-12 and Th1-cytokines play an important role in intestinal inflammation. We therefore examined the role of IL-12 and interferon-gamma (IFN-gamma) in our model of dextran sulfate-induced acute colitis in mice. Treatment of mice with rmIL-12 during colitis induction resulted in severe aggravation as demonstrated by a greater loss of body weight, an increase of the histological parameters, and reduction of myeloperoxidase activity in colonic biopsies. Depletion of neutrophils in mice also led to aggravation of colitis. Neutralization of IFN-gamma in IL-12-treated mice with colitis inhibited these effects of IL-12. Neutralization of endogenous IFN-gamma or IL-12 with specific antibodies in DSS-treated mice, however, had only weak ameliorating effects. Since IL-12 and IFN-gamma have been shown to mediate experimental chronic colitis we conclude that the transition from a macrophage/neutrophil determined response to a Th-cell response promotes chronic intestinal inflammation.


Assuntos
Colite/imunologia , Inflamação/imunologia , Interferon gama/fisiologia , Interleucina-12/toxicidade , Mucosa Intestinal/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana , Feminino , Regulação da Expressão Gênica/imunologia , Inflamação/induzido quimicamente , Inflamação/patologia , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos BALB C , Peroxidase/metabolismo , RNA Mensageiro/genética , Proteínas Recombinantes/farmacologia , Transcrição Gênica , Redução de Peso/efeitos dos fármacos , Redução de Peso/imunologia
20.
Arch Immunol Ther Exp (Warsz) ; 45(1): 49-54, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9090440

RESUMO

Production of interleukin 12 (IL-12) during cytotoxic reaction of C57BL/6 and BALB/c spleen lymphocytes or their subpopulations: T+B, T, CD4+ and CD8+ cells to L. innocua-phagocyting syngeneic macrophages was examined. The effector cell donors were untreated or L. innocua-infected. The number of surviving bacteria in phagocytes was tested and IL-12 level in culture supernatants of reacting cells was determined. C57BL/6 mice, resistant to Listeria infection, were found to develop stronger cell cytotoxicity to bacteria-phagocyting syngeneic macrophages than BALB/c mice. The lymphocytes responsible for that phenomenon were of CD8+ phenotype. IL-12 was produced only during nonspecific cytotoxic reaction of CD4+ and CD8+ T cells. It is suggested that the innate resistance of mice to Listeria is dependent on their ability to develop a specific cytotoxic reaction and IL-12 production.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica/imunologia , Interleucina-12/biossíntese , Interleucina-12/fisiologia , Listeria/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Baço/imunologia , Animais , Linfócitos T CD8-Positivos/microbiologia , Células Cultivadas , Feminino , Imunidade Inata , Interleucina-12/toxicidade , Listeria/crescimento & desenvolvimento , Listeriose/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA