Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
JAMA Oncol ; 9(12): 1660-1668, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37824131

RESUMEN

Importance: Metastatic soft tissue sarcomas (STSs) have limited systemic therapy options, and immunomodulation has not yet meaningfully improved outcomes. Intratumoral (IT) injection of the toll-like receptor 4 (TLR4) agonist glycopyranosyl lipid A in stable-emulsion formulation (GLA-SE) has been studied as immunotherapy in other contexts. Objective: To evaluate the safety, efficacy, and immunomodulatory effects of IT GLA-SE with concurrent radiotherapy in patients with metastatic STS with injectable lesions. Design, Setting, and Participants: This phase 1 nonrandomized controlled trial of patients with STS was performed at a single academic sarcoma specialty center from November 17, 2014, to March 16, 2016. Data analysis was performed from August 2016 to September 2022. Interventions: Two doses of IT GLA-SE (5 µg and 10 µg for 8 weekly doses) were tested for safety in combination with concurrent radiotherapy of the injected lesion. Main Outcomes and Measures: Primary end points were safety and tolerability. Secondary and exploratory end points included local response rates as well as measurement of antitumor immunity with immunohistochemistry and T-cell receptor (TCR) sequencing of tumor-infiltrating and circulating lymphocytes. Results: Twelve patients (median [range] age, 65 [34-78] years; 8 [67%] female) were treated across the 2 dose cohorts. Intratumoral GLA-SE was well tolerated, with only 1 patient (8%) experiencing a grade 2 adverse event. All patients achieved local control of the injected lesion after 8 doses, with 1 patient having complete regression (mean regression, -25%; range, -100% to 4%). In patients with durable local response, there were detectable increases in tumor-infiltrating lymphocytes. In 1 patient (target lesion -39% at 259 days of follow-up), TCR sequencing revealed expansion of preexisting and de novo clonotypes, with convergence of numerous rearrangements coding for the same binding sequence (suggestive of clonal convergence to antitumor targets). Single-cell sequencing identified these same expanded TCR clones in peripheral blood after treatment; these T cells had markedly enhanced Tbet expression, suggesting TH1 phenotype. Conclusions and Relevance: In this nonrandomized controlled trial, IT GLA-SE with concurrent radiotherapy was well tolerated and provided more durable local control than radiotherapy alone. Patients with durable local response demonstrated enhanced IT T-cell clonal expansion, with matched expansion of these clonotypes in the circulation. Additional studies evaluating synergism of IT GLA-SE and radiotherapy with systemic immune modulation are warranted. Trial Registration: ClinicalTrials.gov Identifier: NCT02180698.


Asunto(s)
Sarcoma , Neoplasias de los Tejidos Blandos , Humanos , Femenino , Anciano , Masculino , Receptor Toll-Like 4/agonistas , Linfocitos T , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/radioterapia , Sarcoma/tratamiento farmacológico , Sarcoma/radioterapia , Receptores de Antígenos de Linfocitos T
2.
Sci Rep ; 13(1): 4648, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36944687

RESUMEN

SARS-CoV-2 continues to circulate in the human population necessitating regular booster immunization for its long-term control. Ideally, vaccines should ideally not only protect against symptomatic disease, but also prevent transmission via asymptomatic shedding and cover existing and future variants of the virus. This may ultimately only be possible through induction of potent and long-lasting immune responses in the nasopharyngeal tract, the initial entry site of SARS-CoV-2. To this end, we have designed a vaccine based on recombinantly expressed receptor binding domain (RBD) of SARS-CoV-2, fused to the C-terminus of C. perfringens enterotoxin, which is known to target Claudin-4, a matrix molecule highly expressed on mucosal microfold (M) cells of the nasal and bronchial-associated lymphoid tissues. To further enhance immune responses, the vaccine was adjuvanted with a novel toll-like receptor 3/RIG-I agonist (Riboxxim™), consisting of synthetic short double stranded RNA. Intranasal prime-boost immunization of mice induced robust mucosal and systemic anti-SARS-CoV-2 neutralizing antibody responses against SARS-CoV-2 strains Wuhan-Hu-1, and several variants (B.1.351/beta, B.1.1.7/alpha, B.1.617.2/delta), as well as systemic T-cell responses. A combination vaccine with M-cell targeted recombinant HA1 from an H1N1 G4 influenza strain also induced mucosal and systemic antibodies against influenza. Taken together, the data show that development of an intranasal SARS-CoV-2 vaccine based on recombinant RBD adjuvanted with a TLR3 agonist is feasible, also as a combination vaccine against influenza.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Gripe Humana , Animales , Humanos , Ratones , Adyuvantes Inmunológicos , Adyuvantes Farmacéuticos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Clostridium perfringens , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Mucosa Gástrica , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Células M , SARS-CoV-2 , Receptor Toll-Like 3
3.
PLoS One ; 16(12): e0259301, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855754

RESUMEN

Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.


Asunto(s)
Glucósidos/farmacología , Memoria Inmunológica/efectos de los fármacos , Interleucina-12/genética , Lípido A/farmacología , Neoplasias Experimentales/inmunología , Receptor Toll-Like 4/agonistas , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Memoria Inmunológica/genética , Inmunoterapia/métodos , Interferón gamma/sangre , Interleucina-12/sangre , Interleucina-12/inmunología , Lentivirus/genética , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología
4.
Nanomaterials (Basel) ; 11(2)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572813

RESUMEN

Bio-inspired surfaces with superamphiphobic properties are well known as effective candidates for antifouling technology. However, the limitation of large-area mastering, patterning and pattern collapsing upon physical contact are the bottleneck for practical utilization in marine and medical applications. In this study, a roll-to-plate nanoimprint lithography (R2P NIL) process using Morphotonics' automated Portis NIL600 tool was used to replicate high aspect ratio (5.0) micro-structures via reusable intermediate flexible stamps that were fabricated from silicon master molds. Two types of Morphotonics' in-house UV-curable resins were used to replicate a micro-pillar (PIL) and circular rings with eight stripe supporters (C-RESS) micro-structure onto polycarbonate (PC) and polyethylene terephthalate (PET) foil substrates. The pattern quality and surface wettability was compared to a conventional polydimethylsiloxane (PDMS) soft lithography process. It was found that the heights of the R2P NIL replicated PIL and C-RESS patterns deviated less than 6% and 5% from the pattern design, respectively. Moreover, the surface wettability of the imprinted PIL and C-RESS patterns was found to be superhydro- and oleophobic and hydro- and oleophobic, respectively, with good robustness for the C-RESS micro-structure. Therefore, the R2P NIL process is expected to be a promising method to fabricate robust C-RESS micro-structures for large-scale anti-biofouling application.

5.
Oncoimmunology ; 9(1): 1847846, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33312760

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Sarcoma , Adyuvantes Inmunológicos , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/efectos adversos , Humanos , Proteínas de la Membrana/genética
6.
Front Oncol ; 10: 1438, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32974162

RESUMEN

Intratumoral (IT) injections of Glucopyranosyl lipid A (G100), a synthetic toll-like receptor 4 (TLR4) agonist formulated in a stable emulsion, resulted in T-cell inflammation of the tumor microenvironment (TME) and complete cure of 60% of mice with large established A20 lymphomas. Strong abscopal effects on un-injected lesions were observed in a bilateral tumor model and surviving mice resisted a secondary tumor challenge. Depletion of CD8 T-cells, but not CD4 or NK cells, abrogated the anti-tumor effect. Unexpectedly, TLR4 knock-out rendered A20 tumors completely non-responsive to G100. In vitro studies showed that GLA has direct effect on A20 cells, but not on A20 cells deficient for TLR4. As shown by genotyping and phenotyping analysis, G100 strongly activated antigen presentation functions in A20 cells in vitro and in vivo and induced their apoptosis in a dose dependent manner. Similarly, the TLR4 positive human mantle cell lymphoma line Mino showed in vitro activation with G100 that was blocked with an anti-TLR4 antibody. In the A20 model, direct activation of B-lymphoma cells with G100 is sufficient to induce protective CD8 T-cell responses and TLR4 expressing human B-cell lymphomas may be amenable to this therapy as well.

7.
Vaccine ; 38(41): 6367-6373, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32828576

RESUMEN

Therapeutic cancer vaccines must induce high levels of tumor-specific cytotoxic CD8 T cells to be effective. We show here that tumor-antigen specific effector and memory T cell responses primed with a non-integrating, dendritic-cell targeted lentiviral vector (ZVex™) could be boosted significantly by either adjuvanted recombinant protein, adenoviral vectors, or self-replicating RNA. These heterologous prime-boost regimens also provided significantly better protection in murine tumor models. In contrast, homologous prime-boost regimens, or using the lentiviral vector as a boost, resulted in lower T cell responses with limited therapeutic efficacy. Heterologous prime-boost regimens that utilize ZVex as the prime may be attractive modalities for therapeutic cancer vaccines.


Asunto(s)
Vacunas de ADN , Vacunas Virales , Adyuvantes Inmunológicos , Animales , Linfocitos T CD8-positivos , Vectores Genéticos , Inmunización Secundaria , Ratones
8.
NPJ Vaccines ; 5(1): 50, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32579133

RESUMEN

Effective T cell-based immunotherapy of solid malignancies requires intratumoral activity of cytotoxic T cells and induction of protective immune memory. A major obstacle to intratumoral trafficking and activation of vaccine-primed or adoptively transferred tumor-specific T cells is the immunosuppressive tumor microenvironment (TME), which currently limits the efficacy of both anti-tumor vaccines and adoptive cell therapy (ACT). Combination treatments to overcome TME-mediated immunosuppression are therefore urgently needed. We combined intratumoral administration of the synthetic toll-like receptor 4 agonist glucopyranosyl lipid A (oil-in-water formulation, G100) with either active vaccination or adoptive transfer of tumor-specific CD8 T cells to mice bearing established melanomas or orthotopically inoculated glioblastomas. In combination with cancer vaccines or ACT, G100 significantly increased expression of innate immune genes, infiltration and expansion of activated effector T cells, antigen spreading, and durable immune responses. Complete tumor regression of both injected and non-injected tumors was observed only in mice receiving combination immunotherapy. TLR4-based intratumoral immune activation may be a viable approach to enhance the efficacy of therapeutic cancer vaccines and ACT in patients.

9.
Vaccine ; 38(17): 3369-3377, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32088020

RESUMEN

While immune checkpoint inhibition is rapidly becoming standard of care in many solid tumors, immune checkpoint inhibitors (ICIs) fail to induce clinical responses in many patients, presumably due to insufficient numbers of tumor-specific T cells in the tumor milieu. To this end, immunization protocols using viral vectors expressing tumor-associated antigens are being explored to induce T cell responses that synergize with ICIs. However, the optimal combination of vaccine and immune checkpoint regimen remains undefined. Here, a dendritic cell-targeting lentiviral vector (ZVex®) expressing the endogenous murine tyrosinase-related protein 1 (mTRP1), or the human tumor antigen NY-ESO-1, was explored as monotherapy or heterologous prime-boost (HPB) vaccine regimen together with recombinant tumor antigen in the murine B16 melanoma model. PD1/PDL1 blockade significantly enhanced ZVex/mTRP1, but not ZVex/NY-ESO-1, induced immune responses in mice, whereas the opposite effect was observed with anti-CTLA4 antibody. Anti-tumor efficacy of anti-PD1, but not anti-PDL1 or anti-CTLA4, was significantly enhanced by ZVex/mTRP1 and HPB vaccination. These results suggest mechanistic differences in the effect of checkpoint blockade on vaccine-induced immune and anti-tumor responses against self versus non-self tumor antigens, possibly due to tolerance and state of exhaustion of anti-tumor T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Melanoma Experimental , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/inmunología , Oxidorreductasas/inmunología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Vectores Genéticos , Humanos , Lentivirus , Melanoma Experimental/terapia , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
11.
Clin Cancer Res ; 25(19): 5808-5817, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31227504

RESUMEN

PURPOSE: LV305 is a modified, third-generation, nonreplicating, integration-deficient lentivirus-based vector designed to selectively transduce dendritic cells in vivo. LV305 induces expression of the New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1) cancer testis antigen in dendritic cells, promoting immune responses against NY-ESO-1-expressing tumors. This phase I study evaluated the safety, immunogenicity, and preliminary efficacy of LV305 in patients with sarcoma or other solid tumors. PATIENTS AND METHODS: Adults with previously treated, advanced, NY-ESO-1-positive solid tumors and limited tumor burden were eligible. LV305 was administered every 3 weeks by intradermal injection in four dose cohorts (Cohort 1: 108 vector genomes (vg) x 3 doses; Cohorts 1A, 2, and 3: 108 vg, 109 vg, 1010 vg x 4 doses). RESULTS: Thirty-nine patients were enrolled: 3 patients each in Cohorts 1, 1A, and 2, and 30 patients in Cohort 3. No dose-limiting toxicities were observed. Tumor types included sarcoma (n = 24), ovarian (n = 8), melanoma (n = 6), and lung cancer (n = 1). All treatment-related adverse events were grade 1 or 2. Common treatment-related adverse events were fatigue (49%), injection site reactions (46%), and myalgia (21%). The disease control rate was 56.4% in all patients and 62.5% in sarcoma patients. One patient with synovial sarcoma achieved a partial response lasting >36 months. Anti-NY-ESO-1-specific CD4+ and/or CD8+ T cells were induced in 57% of evaluable sarcoma patients. Induction of an anti-NY-ESO-1 immune response was associated with improved 1-year survival in an exploratory analysis. CONCLUSIONS: This first-in-class, first-in-human study of LV305 demonstrated a favorable safety profile, induction of antigen-specific responses, and potential clinical activity in patients with advanced cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/trasplante , Vectores Genéticos/administración & dosificación , Lentivirus/genética , Proteínas de la Membrana/inmunología , Neoplasias/terapia , Sarcoma/terapia , Adulto , Anciano , Antígenos de Neoplasias/biosíntesis , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Estudios de Cohortes , Células Dendríticas/citología , Células Dendríticas/inmunología , Supervivencia sin Enfermedad , Femenino , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Humanos , Lentivirus/inmunología , Masculino , Proteínas de la Membrana/biosíntesis , Persona de Mediana Edad , Neoplasias/inmunología , Sarcoma/inmunología , Tasa de Supervivencia
12.
Cancer Immunol Immunother ; 68(7): 1211-1222, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31069460

RESUMEN

Human tumor cells express antigens that serve as targets for the host cellular immune system. This phase 1 dose-escalating study was conducted to assess safety and tolerability of G305, a recombinant NY-ESO-1 protein vaccine mixed with glucopyranosyl lipid A (GLA), a synthetic TLR4 agonist adjuvant, in a stable emulsion (SE). Twelve patients with solid tumors expressing NY-ESO-1 were treated using a 3 + 3 design. The NY-ESO-1 dose was fixed at 250 µg, while GLA-SE was increased from 2 to 10 µg. Safety, immunogenicity, and clinical responses were assessed prior to, during, and at the end of therapy. G305 was safe and immunogenic at all doses. All related AEs were Grade 1 or 2, with injection site soreness as the most commonly reported event (100%). Overall, 75% of patients developed antibody response to NY-ESO-1, including six patients with increased antibody titer ( ≥ 4-fold rise) and three patients with seroconversion from negative (titer < 100) to positive (titer ≥ 100). CD4 T-cell responses were observed in 44.4% of patients; 33.3% were new responses and 1 was boosted ( ≥ 2-fold rise). Following treatment, 8 of 12 patients had stable disease for 3 months or more; at the end of 1 year, three patients had stable disease and nine patients were alive. G305 is a potent immunotherapeutic agent that can stimulate NY-ESO-1-specific antibody and T-cell responses. The vaccine was safe at all doses of GLA-SE (2-10 µg) and showed potential clinical benefit in this population of patients.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Glucósidos/administración & dosificación , Lípido A/administración & dosificación , Proteínas de la Membrana/administración & dosificación , Neoplasias/terapia , Adyuvantes Inmunológicos/efectos adversos , Adulto , Anciano , Antígenos de Neoplasias/efectos adversos , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Drogas en Investigación/administración & dosificación , Drogas en Investigación/efectos adversos , Femenino , Glucósidos/efectos adversos , Glucósidos/inmunología , Humanos , Inmunogenicidad Vacunal , Inyecciones Intramusculares , Lípido A/efectos adversos , Lípido A/inmunología , Masculino , Proteínas de la Membrana/efectos adversos , Proteínas de la Membrana/inmunología , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Adulto Joven
13.
Cell Rep ; 26(2): 438-446.e5, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30625326

RESUMEN

Yellow fever virus (YFV), a deadly human pathogen, is the prototype of the genus Flavivirus. Recently, YFV re-emerged in Africa and Brazil, leading to hundreds of deaths, with some cases imported to China. Prophylactic or therapeutic countermeasures are urgently needed. Previously, several human monoclonal antibodies against YFV were screened out by phage display. Here, we find that one of them, 5A, exhibits high neutralizing potency and good protection. Crystallographic analysis of the YFV envelope (E) protein in its pre- and post-fusion states shows conformations similar to those observed in other E proteins of flaviviruses. Furthermore, the structures of 5A in complex with the E protein in both states are resolved, revealing an invariant recognition site. Structural analysis and functional data suggest that 5A has high neutralization potency because it interferes with virus entry by preventing both virus attachment and fusion. These findings will be instrumental for immunogen or inhibitor design.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Simulación del Acoplamiento Molecular , Proteínas del Envoltorio Viral/inmunología , Fiebre Amarilla/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/uso terapéutico , Afinidad de Anticuerpos , Chlorocebus aethiops , Cricetinae , Cricetulus , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Células Vero , Proteínas del Envoltorio Viral/química , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/inmunología
14.
Clin Cancer Res ; 25(4): 1185-1195, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30093453

RESUMEN

PURPOSE: G100 is a toll-like receptor 4 (TLR4) agonist that triggers innate and adaptive antitumor immune responses in preclinical models. This pilot study assessed the safety, efficacy, and immunologic activity of intratumoral (IT) administration of G100 in patients with Merkel cell carcinoma (MCC). PATIENTS AND METHODS: Patients with locoregional MCC (n = 3; cohort A) received neoadjuvant IT G100 (2 weekly doses at 5 µg/dose) followed by surgery and radiotherapy; patients with metastatic MCC (n = 7; cohort B) received 3 doses in a 6-week cycle and could receive additional cycles with/without radiotherapy. RESULTS: IT G100 was safe and feasible in both neoadjuvant and metastatic settings. Treatment-related adverse events were mostly grade 1 or 2 injection-site reactions. IT G100 led to increased inflammation in the injected tumors with infiltration of CD8+ and CD4+ T cells and activation of immune-related genes. These proinflammatory changes were associated with local tumor regression and appeared to promote systemic immunity. All 3 cohort A patients successfully completed therapy; 2 patients remain recurrence free at 44+ and 41+ months, including 1 with a pathologic complete response after G100 alone. In cohort B, 2 patients achieved sustained partial responses, both lasting 33+ months after 2 cycles of therapy. CONCLUSIONS: In this first-in-human study, IT G100 induced antitumor immune responses, demonstrated acceptable safety, and showed encouraging clinical activity.See related commentary by Marquez-Rodas et al., p. 1127.


Asunto(s)
Carcinoma de Células de Merkel/tratamiento farmacológico , Inmunoterapia , Lípido A/análogos & derivados , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor Toll-Like 4/agonistas , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/administración & dosificación , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/inmunología , Carcinoma de Células de Merkel/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Lípido A/farmacología , Lípido A/uso terapéutico , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Proyectos Piloto , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Microambiente Tumoral/efectos de los fármacos
15.
NPJ Vaccines ; 3: 3, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29387473

RESUMEN

The hemagglutinination inhibition (HI) response remains the gold standard used for the licensure of influenza vaccines. However, cell-mediated immunity (CMI) deserves more attention, especially when evaluating H5N1 influenza vaccines that tend to induce poor HI response. In this study, we measured the humoral response (HI) and CMI (flow cytometry) during a Phase II dose-ranging clinical trial (NCT01991561). Subjects received two intramuscular doses, 21 days apart, of plant-derived virus-like particles (VLP) presenting the A/Indonesia/05/2005 H5N1 influenza hemagglutinin protein (H5) at the surface of the VLP (H5VLP). The vaccine was co-administrated with Alhydrogel® or with a glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE). We demonstrated that low doses (3.75 or 7.5 µg H5VLP) of GLA-SE-adjuvanted vaccines induced HI responses that met criteria for licensure at both antigen doses tested. Alhydrogel adjuvanted vaccines induced readily detectable HI response that however failed to meet licensure criteria at any of three doses (10, 15 and 20 µg) tested. The H5VLP also induced a sustained (up to 6 months) polyfunctional and cross-reactive HA-specific CD4+ T cell response in all vaccinated groups. Interestingly, the frequency of central memory Th1-primed precursor cells before the boost significantly correlated with HI titers 21 days after the boost. The ability of the low dose GLA-SE-adjuvanted H5VLP to elicit both humoral response and a sustained cross-reactive CMI in healthy adults is very attractive and could result in significant dose-sparing in a pandemic situation.

16.
J Immunother ; 40(8): 302-306, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28891906

RESUMEN

Effective induction of antitumor T cells is a pivotal goal of cancer immunotherapy. To this end, lentiviral vectors (LV) are uniquely poised to directly prime CD8 T-cell responses via transduction of dendritic cells in vivo and have shown promise as active cancer therapeutics in preclinical tumor models. However, until now, significant barriers related to production and regulation have prevented their widespread use in the clinic. We developed LV305, a dendritic cell-targeting, integration-deficient, replication incompetent LV from the ZVex platform, encoding the full-length cancer-testis antigen NY-ESO-1. LV305 is currently being evaluated in phase 1 and 2 trials in metastatic recurrent cancer patients with NY-ESO-1 positive solid tumors as a single agent and in combination with anti-PD-L1. Here we report on the first patient treated with LV305, a young woman with metastatic, recurrent, therapy-refractive NY-ESO-1 synovial sarcoma. The patient developed a robust NY-ESO-1-specific CD4 and CD8 T-cell response after 3 intradermal injections with LV305, and subsequently over 85% disease regression that is continuing for >2.5 years posttherapy. No adverse events >grade 2 occurred. This case demonstrates that LV305 can be safely administered and has the potential to induce a significant clinical benefit and immunologic response in a patient with advanced stage cancer.


Asunto(s)
Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Sarcoma Sinovial/terapia , Adulto , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células Dendríticas/trasplante , Resistencia a Antineoplásicos , Femenino , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neumonectomía , Receptor de Muerte Celular Programada 1/metabolismo , Recurrencia , Inducción de Remisión , Sarcoma Sinovial/inmunología , Sarcoma Sinovial/secundario
17.
Mol Ther ; 25(2): 494-503, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28153096

RESUMEN

To be effective against HIV type 1 (HIV-1), vaccine-induced T cells must selectively target epitopes, which are functionally conserved (present in the majority of currently circulating and reactivated HIV-1 strains) and, at the same time, beneficial (responses to which are associated with better clinical status and control of HIV-1 replication), and rapidly reach protective frequencies upon exposure to the virus. Heterologous prime-boost regimens using virally vectored vaccines are currently the most promising vaccine strategies; nevertheless, induction of robust long-term memory remains challenging. To this end, lentiviral vectors induce high frequencies of memory cells due to their low-inflammatory nature, while typically inducing only low anti-vector immune responses. Here, we describe construction of novel candidate vaccines ZVex.tHIVconsv1 and ZVex.tHIVconsv2, which are based on an integration-deficient lentiviral vector platform with preferential transduction of human dendritic cells and express a bivalent mosaic of conserved-region T cell immunogens with a high global HIV-1 match. Each of the two mosaic vaccines was individually immunogenic. When administered together in heterologous prime-boost regimens with chimpanzee adenovirus and/or poxvirus modified vaccinia virus Ankara (MVA) vaccines to BALB/c and outbred CD1-Swiss mice, they induced a median frequency of over 6,000 T cells/106 splenocytes, which were plurifunctional, broadly specific, and cross-reactive. These results support further development of this vaccine concept.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vectores Genéticos/genética , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/inmunología , Lentivirus/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Animales , Secuencia Conservada , Modelos Animales de Enfermedad , Epítopos/genética , Epítopos/inmunología , Femenino , Orden Génico , Infecciones por VIH/virología , Humanos , Inmunidad Celular , Ratones , Péptidos/genética , Péptidos/inmunología
18.
Hum Vaccin Immunother ; 13(1): 15-16, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870594
19.
Mol Ther Oncolytics ; 3: 16010, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27626061

RESUMEN

We have engineered an integration-deficient lentiviral vector, LV305, to deliver the tumor antigen NY-ESO-1 to human dendritic cells in vivo through pseudotyping with a modified Sindbis virus envelop protein. Mice immunized once with LV305 developed strong, dose-dependent, multifunctional, and cytotoxic NY-ESO-1-specific cluster of differentiation 8 (CD8) T cells within 14 days post-immunization and could be boosted with LV305 at least twice to recall peak-level CD8 T-cell responses. Immunization with LV305 protected mice against tumor growth in an NY-ESO-1-expressing CT26 lung metastasis model, with the protective effect abrogated upon depletion of CD8 T cells. Adoptive transfer of CD8 T cells, alone or together with CD4 T cells or natural killer cells, from LV305-immunized donor mice to tumor-bearing recipient mice conferred significant protection against metastatic tumor growth. Biodistribution of injected LV305 in mice was limited to the site of injection and the draining lymph node, and injected LV305 exhibited minimal excretion. Mice injected with LV305 developed little to no adverse effects, as evaluated by toxicology studies adherent to good laboratory practices. Taken together, these data support the development of LV305 as a clinical candidate for treatment against tumors expressing NY-ESO-1.

20.
J Infect Dis ; 214(suppl 3): S342-S354, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27456709

RESUMEN

The 2013-2016 West African Ebola virus (EBOV) disease outbreak was the largest filovirus outbreak to date. Over 28 000 suspected, probable, or confirmed cases have been reported, with a 53% case-fatality rate. The magnitude and international impact of this EBOV outbreak has highlighted the urgent need for a safe and efficient EBOV vaccine. To this end, we demonstrate the immunogenicity and protective efficacy of FILORAB1, a recombinant, bivalent, inactivated rabies virus-based EBOV vaccine, in rhesus and cynomolgus monkeys. Our results demonstrate that the use of the synthetic Toll-like receptor 4 agonist glucopyranosyl lipid A in stable emulsion (GLA-SE) as an adjuvant increased the efficacy of FILORAB1 to 100% protection against lethal EBOV challenge, with no to mild clinical signs of disease. Furthermore, all vaccinated subjects developed protective anti-rabies virus antibody titers. Taken together, these results support further development of FILORAB1/GLA-SE as an effective preexposure EBOV vaccine.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Glucósidos/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Lípido A/inmunología , Virus de la Rabia/inmunología , Rabia/prevención & control , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales/inmunología , Emulsiones , Femenino , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Macaca fascicularis , Macaca mulatta , Masculino , Rabia/inmunología , Rabia/virología , Vacunas Antirrábicas/inmunología , Receptor Toll-Like 4/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...