Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1362775, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38487528

RESUMO

Background: The benefits of recombinant interleukin-12 (rIL-12) as a multifunctional cytokine and potential immunotherapy for cancer have been sought for decades based on its efficacy in multiple mouse models. Unexpected toxicity in the first phase 2 study required careful attention to revised dosing strategies. Despite some signs of efficacy since then, most rIL-12 clinical trials have encountered hurdles such as short terminal elimination half-life (T½), limited tumor microenvironment targeting, and substantial systemic toxicity. We developed a strategy to extend the rIL-12 T½ that depends on binding albumin in vivo to target tumor tissue, using single-chain rIL-12 linked to a fully human albumin binding (FHAB) domain (SON-1010). After initiating a dose-escalation trial in patients with cancer (SB101), a randomized, double-blind, placebo-controlled, single-ascending dose (SAD) phase 1 trial in healthy volunteers (SB102) was conducted. Methods: SB102 (NCT05408572) focused on safety, tolerability, pharmacokinetic (PK), and pharmacodynamic (PD) endpoints. SON-1010 at 50-300 ng/kg or placebo administered subcutaneously on day 1 was studied at a ratio of 6:2, starting with two sentinels; participants were followed through day 29. Safety was reviewed after day 22, before enrolling the next cohort. A non-compartmental analysis of PK was performed and correlations with the PD results were explored, along with a comparison of the SON-1010 PK profile in SB101. Results: Participants receiving SON-1010 at 100 ng/kg or higher tolerated the injection but generally experienced more treatment-emergent adverse effects (TEAEs) than those receiving the lowest dose. All TEAEs were transient and no other dose relationship was noted. As expected with rIL-12, initial decreases in neutrophils and lymphocytes returned to baseline by days 9-11. PK analysis showed two-compartment elimination in SB102 with mean T½ of 104 h, compared with one-compartment elimination in SB101, which correlated with prolonged but controlled and dose-related increases in interferon-gamma (IFNγ). There was no evidence of cytokine release syndrome based on minimal participant symptoms and responses observed with other cytokines. Conclusion: SON-1010, a novel presentation for rIL-12, was safe and well-tolerated in healthy volunteers up to 300 ng/kg. Its extended half-life leads to a prolonged but controlled IFNγ response, which may be important for tumor control in patients. Clinical trial registration: https://clinicaltrials.gov/study/NCT05408572, identifier NCT05408572.


Assuntos
Interleucina-12 , Neoplasias , Animais , Camundongos , Humanos , Citocinas , Voluntários Saudáveis , Neoplasias/tratamento farmacológico , Interferon gama , Interleucina-2 , Proteínas Recombinantes , Albuminas/efeitos adversos , Microambiente Tumoral
2.
Front Immunol ; 14: 1326927, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38250068

RESUMO

Background: The potential synergy between interleukin-12 (IL-12) and IL-15 holds promise for more effective solid tumor immunotherapy. Nevertheless, previous clinical trials involving therapeutic cytokines have encountered obstacles such as short pharmacokinetics, limited tumor microenvironment (TME) targeting, and substantial systemic toxicity. Methods: To address these challenges, we fused single-chain human IL-12 and native human IL-15 in cis onto a fully human albumin binding (FHAB) domain single-chain antibody fragment (scFv). This novel fusion protein, IL12-FHAB-IL15 (SON-1210), is anticipated to amplify the therapeutic impact of interleukins and combination immunotherapies in human TME. The molecule was studied in vitro and in animal models to assess its pharmacokinetics, potency, functional characteristics, safety, immune response, and efficacy. Results: SON-1210 demonstrated robust binding affinity to albumin and exhibited the anticipated in vitro activity and tumor model efficacy that might be expected based on decades of research on native IL-12 and IL-15. Notably, in the B16F10 melanoma model (a non-immunogenic, relatively "cold" tumor), the murine counterpart of the construct, which had mouse (m) and human (h) cytokine sequences for the respective payloads (mIL12-FHAB-hIL15), outperformed equimolar doses of the co-administered native cytokines in a dose-dependent manner. A single dose caused a marked reduction in tumor growth that was concomitant with increased IFNγ levels; increased Th1, CTL, and activated NK cells; a shift in macrophages from the M2 to M1 phenotype; and a reduction in Treg cells. In addition, a repeat-dose non-human primate (NHP) toxicology study displayed excellent tolerability up to 62.5 µg/kg of SON-1210 administered three times, which was accompanied by the anticipated increases in IFNγ levels. Toxicokinetic analyses showed sustained serum levels of SON-1210, using a sandwich ELISA with anti-IL-15 for capture and biotinylated anti-IL-12 for detection, along with sustained IFNγ levels, indicating prolonged kinetics and biological activity. Conclusion: Collectively, these findings support the suitability of SON-1210 for patient trials in terms of activity, efficacy, and safety, offering a promising opportunity for solid tumor immunotherapy. Linking cytokine payloads to a fully human albumin binding domain provides an indirect opportunity to target the TME using potent cytokines in cis that can redirect the immune response and control tumor growth.


Assuntos
Interleucina-12 , Melanoma , Humanos , Animais , Camundongos , Citocinas , Interleucina-15 , Meia-Vida , Albumina Sérica Humana , Microambiente Tumoral
3.
Oncoimmunology ; 9(1): 1847846, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33312760

RESUMO

Preclinical data suggest that a "prime-boost" vaccine regimen using a target-expressing lentiviral vector for priming, followed by a recombinant protein boost, may be effective against cancer; however, this strategy has not been evaluated in a clinical setting. CMB305 is a prime-boost vaccine designed to induce a broad anti-NY-ESO-1 immune response. It is composed of LV305, which is an NY-ESO-1 expressing lentiviral vector, and G305, a recombinant adjuvanted NY-ESO-1 protein. This multicenter phase 1b, first-in-human trial evaluated CMB305 in patients with NY-ESO-1 expressing solid tumors. Safety was examined in a 3 + 3 dose-escalation design, followed by an expansion with CMB305 alone or in a combination with either oral metronomic cyclophosphamide or intratumoral injections of a toll-like receptor agonist (glucopyranosyl lipid A). Of the 79 patients who enrolled, 81.0% had sarcomas, 86.1% had metastatic disease, and 57.0% had progressive disease at study entry. The most common adverse events were fatigue (34.2%), nausea (26.6%), and injection-site pain (24.1%). In patients with soft tissue sarcomas, a disease control rate of 61.9% and an overall survival of 26.2 months (95% CI, 22.1-NA) were observed. CMB305 induced anti-NY-ESO-1 antibody and T-cell responses in 62.9% and 47.4% of patients, respectively. This is the first trial to test a prime-boost vaccine regimen in patients with advanced cancer. This approach is feasible, can be delivered safely, and with evidence of immune response as well as suggestion of clinical benefit.


Assuntos
Vacinas Anticâncer , Sarcoma , Adjuvantes Imunológicos , Antígenos de Neoplasias/genética , Vacinas Anticâncer/efeitos adversos , Humanos , Proteínas de Membrana/genética
4.
Am J Trop Med Hyg ; 98(3): 849-856, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29363446

RESUMO

We conducted an open label, dose escalation Phase 1 clinical trial of a tetravalent dengue DNA vaccine (TVDV) formulated in Vaxfectin® to assess safety and immunogenicity. A total of 40 dengue- and flavivirus-naive volunteers received either low-dose (1 mg) TVDV alone (N = 10, group 1), low-dose TVDV (1 mg) formulated in Vaxfectin (N = 10, group 2), or high-dose TVDV (2 mg, group 3) formulated in Vaxfectin® (N = 20). Subjects were immunized intramuscularly with three doses on a 0-, 30-, 90-day schedule and monitored. Blood samples were obtained after each immunization and various time points thereafter to assess anti-dengue antibody and interferon gamma (IFNγ) T-cell immune responses. The most common adverse events (AEs) across all groups included mild to moderate pain and tenderness at the injection site, which typically resolved within 7 days. Common solicited signs and symptoms included fatigue (42.5%), headache (45%), and myalgias (47.5%). There were no serious AEs related to the vaccine or study procedures. No anti-dengue antibody responses were detected in group 1 subjects who received all three immunizations. There were minimal enzyme-linked immunosorbent assay and neutralizing antibody responses among groups 2 and 3 subjects who completed the immunization schedule. By contrast, IFNγ T-cell responses, regardless of serotype specificity, occurred in 70%, 50%, and 79% of subjects in groups 1, 2, and 3, respectively. The largest IFNγ T-cell responses were among group 3 subjects. We conclude that TVDV was safe and well-tolerated and elicited predominately anti-dengue T-cell IFNγ responses in a dose-related fashion.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Vacinas contra Dengue/administração & dosagem , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Imunidade Celular/efeitos dos fármacos , Vacinas de DNA/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Adulto , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/efeitos adversos , Fadiga/etiologia , Fadiga/fisiopatologia , Feminino , Cefaleia/etiologia , Cefaleia/fisiopatologia , Humanos , Esquemas de Imunização , Imunogenicidade da Vacina , Injeções Intramusculares , Interferon gama/biossíntese , Interferon gama/imunologia , Masculino , Mialgia/etiologia , Mialgia/fisiopatologia , Segurança do Paciente , Fosfatidiletanolaminas/administração & dosagem , Fosfatidiletanolaminas/química , Vacinação , Vacinas de DNA/efeitos adversos
6.
Lancet Infect Dis ; 12(4): 290-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22237175

RESUMO

BACKGROUND: Cytomegalovirus reactivation occurs within 6 months in 60-70% of cytomegalovirus-seropositive patients after allogeneic haemopoietic stem-cell transplantation (HSCT), mainly due to immunosuppression associated with the procedure. Pre-emptive antiviral therapy reduces incidence of cytomegalovirus disease but can be toxic. To reduce the potential for disease and subsequent need for such antiviral drugs, we aimed to assess safety and efficacy of a cytomegalovirus therapeutic DNA vaccine compared with placebo. METHODS: In this exploratory double-blind, placebo-controlled, parallel group, phase 2 trial, up to 80 donor-recipient pairs and 80 unpaired recipients undergoing allogeneic HSCT were planned for enrolment at 16 transplant centres in the USA. Eligible recipients were cytomegalovirus-seropositive, 18-65 years old, without high-risk primary disease, T-cell depletion, previous vaccination for cytomegalovirus, or autoimmune diseases. We randomly allocated participants in both parallel groups in a 1:1 ratio to receive a cytomegalovirus therapeutic DNA vaccine (TransVax; Vical, San Diego, CA, USA) or placebo before conditioning and at 1, 3, and 6 months after transplantation. The vaccine contains plasmids encoding cytomegalovirus glycoprotein B and phosphoprotein 65 formulated with poloxamer CRL1005 and benzalkonium chloride. Randomisation was done by sequential allocation based on Pocock and Simon's method, and stratified by site, donor-recipient HLA matching status, and donor's cytomegalovirus serostatus. The primary outcome was the occurrence rate of clinically significant viraemia resulting in initiation of cytomegalovirus-specific antiviral therapy in the per-protocol assessable population. We assessed rates of adverse events in all participants who received at least one dose of vaccine or placebo. This study is registered with ClinicalTrials.gov, number NCT00285259. FINDINGS: We randomly allocated 108 participants (94 HSCT recipients and 14 paired donors) between June 29, 2006, and Dec 11, 2009. Enrolment of the paired arm was halted in February 2008 for logistical reasons. Safety was assessed in all participants; the efficacy population was restricted to 74 unpaired recipients. Groups were balanced for demographic and clinical variables. 19 (48%) of 40 vaccine recipients required cytomegalovirus-specific antiviral therapy, compared with 21 (62%) of 34 controls (p=0·145). However, during follow-up vaccine significantly reduced the occurrence and recurrence of cytomegalovirus viraemia and improved the time-to-event for viraemia episodes compared with placebo. The vaccine was well-tolerated; only one participant discontinued after an allergic reaction. Incidence of common adverse events after HSCT (eg, graft-versus-host disease or secondary infections) did not differ between groups. INTERPRETATION: We show proof of concept for an immunotherapeutic cytomegalovirus vaccine (TransVax) for clinically significant viraemia in the HSCT setting. The reported safety and efficacy outcomes support further development in a phase 3 trial, notwithstanding a lack of significant reduction in the use of cytomegalovirus-specific antiviral therapy compared with placebo in this phase 2 trial. FUNDING: Vical and US National Institute of Allergy and Infectious Diseases.


Assuntos
Infecções por Citomegalovirus/terapia , Vacinas contra Citomegalovirus/administração & dosagem , Citomegalovirus/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Vacinas de DNA/administração & dosagem , Adulto , Idoso , Anticorpos Antivirais/sangue , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/prevenção & controle , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/imunologia , Método Duplo-Cego , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Viremia/imunologia , Viremia/terapia , Viremia/virologia , Adulto Jovem
7.
Vaccine ; 25(34): 6334-40, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17640780

RESUMO

The feasibility of developing a prophylactic vaccine against SARS was assessed by comparing the immune responses elicited by immunizing mice with a recombinant SARS spike glycoprotein (S-protein) formulated with different adjuvants, given by different routes. In both young and aged mice, an intranasal Protollin-formulated S-protein vaccine elicited high levels of antigen-specific IgG in serum, comparable to those elicited by an intramuscular Alum-adsorbed S-protein vaccine. Serum antibodies were shown to be virus neutralizing. Intranasal immunization of young mice with the Protollin-formulated vaccine elicited significant levels of antigen-specific lung IgA in contrast to mice immunized with the intramuscular vaccine in which no antigen-specific lung IgA was detected. Following live virus challenge of aged mice, no virus was detected in the lungs of intranasally immunized mice, in contrast to intramuscularly immunized mice whose lung virus titers were comparable to those observed in control mice.


Assuntos
Anticorpos Antivirais/biossíntese , Cisteína Endopeptidases/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Pulmão/imunologia , Glicoproteínas de Membrana/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Administração Intranasal , Animais , Cisteína Endopeptidases/imunologia , Citocinas/biossíntese , Combinação de Medicamentos , Feminino , Imunização , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia
8.
Vaccine ; 23(7): 946-50, 2005 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-15603897

RESUMO

The elderly have greater morbidity and mortality due to influenza, and respond poorly to influenza vaccination compared to younger adults. This study was designed to determine if the adjuvant heat-labile enterotoxin from Escherichia coli (LT), administered as an immunostimulant (IS) patch on the skin with influenza vaccination, improves influenza immune responses in the elderly. Three weeks following vaccination, hemagglutination inhibition (HAI) responses in LT IS patch recipients showed improvement over those of elderly receiving vaccine alone, as demonstrated by significance or trends in fold rise [A/Panama (P = 0.004), A/New Caledonia (P = 0.09)], seroconversion [A/New Caledonia (63% versus 40%, P = 0.01), A/Panama (54% versus 36%, P = 0.08)] and seroprotection [26%, 20% and 16% greater for the patch group for A/New Caledonia, A/Panama and B/Shandong strains, respectively]. The data suggest that an LT IS patch may further enhance influenza vaccine immune responses in the elderly.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Idoso , Toxinas Bacterianas/administração & dosagem , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Administração Cutânea , Adolescente , Adulto , Toxinas Bacterianas/efeitos adversos , Toxinas Bacterianas/imunologia , Enterotoxinas/efeitos adversos , Enterotoxinas/imunologia , Proteínas de Escherichia coli/efeitos adversos , Proteínas de Escherichia coli/imunologia , Feminino , Humanos , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Masculino , Pessoa de Meia-Idade
9.
N Engl J Med ; 351(22): 2295-301, 2004 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-15525714

RESUMO

BACKGROUND: The loss of half the U.S. supply of influenza vaccine due to contamination has created a critical shortage. Dose-sparing strategies that use intradermal delivery of vaccines may be one approach to consider. METHODS: We conducted a randomized, open-label trial outside the influenza season in 100 healthy adults 18 to 40 years of age to compare the immunogenicity and safety of intradermal immunization with influenza vaccine with standard intramuscular immunization. Subjects were randomly assigned to receive either a single intramuscular dose of 0.5 ml of trivalent influenza vaccine, containing at least 15 microg of hemagglutinin per strain, by means of a prefilled syringe or a single intradermal dose of 0.1 ml, containing at least 3 microg of hemagglutinin per strain, by means of a fine-gauge needle; both injections were in the deltoid region. Changes in the hemagglutination-inhibition (HAI) antibody titer were assessed by comparing geometric mean titers and fold increases relative to baseline values and by comparing changes in the seroconversion and seroprotection rates. Local and systemic adverse events were assessed after both types of vaccination. RESULTS: Subjects who received an intradermal injection with one fifth the standard dose of influenza vaccine had increases in the geometric mean HAI titer by a factor of 15.2 for the H1N1 strain in the vaccine, 19.0 for the H3N2 strain, and 12.4 for the B strain on day 21, as compared with respective increases by a factor of 14.9, 7.1, and 15.3 for the intramuscular injection of the standard dose. Seroconversion and seroprotection rates were similar in the two groups on day 21, ranging from 66 to 82 percent and 84 to 100 percent, respectively. Local reactions were significantly more frequent among recipients of intradermal injections than among recipients of intramuscular injections, but such reactions were mild and transient. CONCLUSIONS: In this study of young adults, intradermal administration of one fifth the standard intramuscular dose of an influenza vaccine elicited immunogenicity that was similar to or better than that elicited by intramuscular injection. Intradermal administration could be used to expand the supplies of influenza vaccine, but further studies are needed before this strategy can be recommended for routine use.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Adolescente , Adulto , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Vacinas contra Influenza/efeitos adversos , Influenza Humana/imunologia , Injeções Intradérmicas , Injeções Intramusculares , Masculino
10.
J Infect Dis ; 190(4): 774-82, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15272406

RESUMO

BACKGROUND: Transcutaneous immunization (TCI) is a needle-free technique that delivers antigens and adjuvants to potent epidermal immune cells. To address critical unmet needs in biodefense against anthrax, we have designed a novel vaccine delivery system using a dry adhesive patch that simplifies administration and improves tolerability of a subunit anthrax vaccine. METHODS: Mice and rabbits were vaccinated with recombinant protective antigen of Bacillus anthracis and the heat-labile toxin of Escherichia coli. Serologic changes, levels of toxin-neutralizing antibodies (TNAs), and pulmonary and nodal responses were monitored in the mice. A lethal aerosolized B. anthracis challenge model was used in A/J mice, to demonstrate efficacy. RESULTS: The level of systemic immunity and protection induced by TCI was comparable to that induced by intramuscular vaccination, and peak immunity could be achieved with only 2 doses. The addition of adjuvant in the patch induced superior TNA levels, compared with injected vaccination. CONCLUSIONS: Anthrax vaccine patches stimulated robust and functional immune responses that protected against lethal challenge. Demonstration of responses in the lung suggests that a mechanism exists for protection against challenge with aerosolized anthrax spores. A formulated, pressure-sensitive, dry adhesive patch, which is stable and can be manufactured in large scale, elicited comparable immunoglobulin G and TNA responses, suggesting that an anthrax vaccine patch is feasible and should advance into clinical evaluation.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Bacillus anthracis/imunologia , Proteínas de Escherichia coli , Vacinação , Adjuvantes Imunológicos , Administração Cutânea , Animais , Antraz/imunologia , Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Enterotoxinas/administração & dosagem , Enterotoxinas/imunologia , Linfonodos/imunologia , Camundongos , Testes de Neutralização , Coelhos , Proteínas Recombinantes/imunologia , Fatores de Tempo , Vacinas de Subunidades/administração & dosagem , Vacinas de Subunidades/imunologia
11.
Expert Rev Vaccines ; 2(2): 167-88, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12899569

RESUMO

Although there are only four adjuvants used in licensed vaccines for humans, a wealth of information on novel vaccine adjuvants has become available in both animal models and clinical studies over the past decade. Many vaccine candidates require immunopotentiation to achieve a satisfactory immune response, which is driving the search for new and safer approaches. In this review, we take a brief look at what is known of the mechanisms of action, consider some of the elements of product development, then survey several of the classes of adjuvants within the context of human trials.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Animais , Química Farmacêutica , Ensaios Clínicos como Assunto , Humanos , Modelos Animais , Modelos Imunológicos , Segurança , Vacinas/efeitos adversos
12.
Expert Rev Vaccines ; 2(2): 253-67, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12899576

RESUMO

The skin is an attractive target for vaccine delivery. Topical application of adjuvants results in potent immune responses and good safety profiles. Adjuvants can be coadministered in a patch with vaccine antigens (transcutaneous immunization) or similar delivery format, or administered separately with an injection or IS patch (Iomai), leading to enhanced immune responses. These observations have moved into the clinic, highlighting the likelihood that skin delivery of vaccines will play an important future role in vaccine applications.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Pele/imunologia , Vacinas/administração & dosagem , Administração Cutânea , Animais , Cobaias , Humanos , Imunidade nas Mucosas , Imunocompetência , Células de Langerhans/imunologia , Camundongos
13.
Exp Parasitol ; 103(3-4): 160-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12880593

RESUMO

Cutaneous leishmaniasis, a parasitic infection causing ulcerating skin lesions, is an important disease worldwide and urgently requires a vaccine. Animal models that closely mimic human disease are essential for designing preventive vaccines against Leishmania major. We have evaluated both biologic and immunologic parameters of cutaneous L. major infection in nonhuman primates. Naïve rhesus macaques or monkeys previously exposed to L. major were infected with varying doses of L. major metacyclic promastigotes, and lesion size was assessed over a 10-week period. Monkeys previously infected with L. major had much smaller lesions that resolved faster compared with those of naïve monkeys in response to the two higher doses of infection. Moreover, eight of nine naïve monkeys had parasites detected in their lesions during the course of the infection. In addition, the cellular infiltrate within the lesions was qualitatively and quantitatively different in naïve versus previously infected monkeys. Finally, an ELIspot assay determined that the magnitude and kinetics of responses differed between previously infected and naïve monkeys.


Assuntos
Modelos Animais de Doenças , Leishmania major , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/fisiopatologia , Macaca mulatta , Animais , Inflamação/imunologia , Inflamação/fisiopatologia , Interferon gama/biossíntese , Leishmania major/imunologia , Leishmania major/patogenicidade , Leishmaniose Cutânea/parasitologia , Leucócitos Mononucleares/imunologia , Pele/imunologia , Pele/parasitologia , Pele/patologia
14.
J Immunol ; 170(9): 4717-23, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12707351

RESUMO

Oligodeoxynucleotides containing CpG motifs (CpG ODNs) mimic microbial DNA and activate effectors of the innate immune response, which limits the spread of pathogens and promotes an adaptive immune response. CpG ODNs have been shown to protect mice from infection with intracellular pathogens. Unfortunately, CpG motifs that optimally stimulate humans are only weakly active in mice, mandating the use of nonhuman primates to monitor the activity and safety of "human" CpG ODNs in vivo. This study demonstrates that CpG ODN treatment of rhesus macaques significantly reduces the severity of the lesions caused by a challenge with Leishmania: Leishmania superinfection is common in immunocompromised hosts, particularly those infected with HIV. This study shows that PBMCs from HIV-infected subjects respond to stimulation with CpG ODNs. To determine whether CpG ODNs can protect retrovirus-infected primates, SIV-infected macaques were treated with CpG ODNs and then challenged with Leishmania: Both lesion size and parasite load were significantly reduced in the CpG-treated animals. These findings support the clinical development of CpG ODNs as immunoprotective agents in normal and HIV-infected patients.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antiprotozoários/uso terapêutico , Ilhas de CpG/imunologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/prevenção & controle , Oligodesoxirribonucleotídeos/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adulto , Animais , Antiprotozoários/administração & dosagem , Células Cultivadas , Feminino , Infecções por HIV/imunologia , Humanos , Injeções Intradérmicas , Leishmania mexicana/imunologia , Leishmaniose Cutânea/virologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Macaca mulatta , Masculino , Oligodesoxirribonucleotídeos/administração & dosagem , Vacinas Protozoárias/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/parasitologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Vírus da Imunodeficiência Símia/imunologia , Carga Viral
15.
Infect Immun ; 71(4): 2095-101, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12654831

RESUMO

Leishmania donovani protozoan parasites, the causative agent of visceral leishmaniasis, establish an infection partly by interfering with cytokine signaling in the host macrophages. Therefore, we investigated the expression of the suppressor of cytokine signaling (SOCS) genes in human macrophages infected with L. donovani. The expression of SOCS3 mRNA was induced transiently after exposure to live or heat-killed parasites, but not purified lipophosphoglycan, while that of other SOCS genes remained unchanged. SOCS3 gene expression was not dependent on phagocytosis or on cytokines released by L. donovani-infected macrophages, such as interleukin-1beta or tumor necrosis factor alpha. In addition, Leishmania used a different signaling pathway(s) than bacterial lipopolysaccharide to induce SOCS3 mRNA, as indicated by the kinetics of induction and sensitivity to polymyxin B inhibition. Finally, phosphorylation of the STAT1 transcription factor was significantly reduced in L. donovani-infected macrophages and required de novo transcription. The induction of SOCS3 provides a potent inhibitory mechanism by which intracellular microorganisms may suppress macrophage activation and interfere with the host immune response.


Assuntos
Leishmania donovani/patogenicidade , Ativação de Macrófagos , Macrófagos/parasitologia , Proteínas/metabolismo , Proteínas Repressoras , Fatores de Transcrição , Animais , Citocinas/metabolismo , Humanos , Interferon gama/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/parasitologia , Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina
16.
Immunol Allergy Clin North Am ; 23(4): 787-813, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14753392

RESUMO

The skin provides an attractive immune environment for vaccine delivery and a safe and confined anatomic space for the use of potent adjuvants. It has been presumed that LCs as a class of dendritic cells should stimulate potent immune responses when activated by adjuvants, and this theory is beginning to be validated. Progress on simple pretreatment of the skin has led to well-developed, simple-to-use protocols that are not dissimilar from current protocols used to cleanse the skin before injection. Antigen and adjuvant formulation optimization has progressed, leading to phase 2 testing of the technology in formulated, manufacturable patches. Although delivery optimization and product testing is challenging, the major biologic observations underlying TCI and the IS patch have been established clearly in that large protein antigens have been delivered clinically, resulting in robust immune responses in a safe manner. During the next 5 years, the challenge will be to conduct a development program that leads to safe and effective vaccination in the context of specific applications.


Assuntos
Imunidade/imunologia , Imunização/métodos , Pele/imunologia , Vacinas/administração & dosagem , Administração Cutânea , Animais , Humanos
18.
J Immunol ; 168(4): 1659-63, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11823494

RESUMO

Synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs act as immune adjuvants in mice, boosting the humoral and cellular response to coadministered Ags. CpG ODN that stimulate human PBMC are only weakly active in mice. Thus, alternative animal models are needed to monitor the activity and safety of "human" CpG ODN in vivo. This work demonstrates that rhesus macaques recognize and respond to the same CpG motifs that trigger human immune cells. Coadministering CpG ODN with heat-killed Leishmania vaccine provided significantly increased protection of macaques against cutaneous Leishmania infection. These findings indicate that rhesus macaques provide a useful model for studying the in vivo activity of human CpG motifs, and that ODN expressing these motifs act as strong immune adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Leishmania major/imunologia , Leishmaniose Cutânea/prevenção & controle , Oligodesoxirribonucleotídeos/farmacologia , Vacinas Protozoárias , Adjuvantes Imunológicos/efeitos adversos , Animais , Anticorpos Antiprotozoários/biossíntese , Antígenos de Protozoários/imunologia , Células Cultivadas , Citocinas/biossíntese , Feminino , Humanos , Cinética , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/patologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Macaca mulatta , Oligodesoxirribonucleotídeos/efeitos adversos , Vacinas de Produtos Inativados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...