Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 72(2): 287-300, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35829790

RESUMO

The development of therapeutic cancer vaccines remains an active area, although previous approaches have yielded disappointing results. We have built on lessons from previous cancer vaccine approaches and immune checkpoint inhibitor research to develop VBIR, a vaccine-based immunotherapy regimen. Assessment of various technologies led to selection of a heterologous vaccine using chimpanzee adenovirus (AdC68) for priming followed by boosts with electroporation of DNA plasmid to deliver T cell antigens to the immune system. We found that priming with AdC68 rapidly activates and expands antigen-specific T cells and does not encounter pre-existing immunity as occurs with the use of a human adenovirus vaccine. The AdC68 vector does, however, induce new anti-virus immune responses, limiting its use for boosting. To circumvent this, boosting with DNA encoding the same antigens can be done repetitively to augment and maintain vaccine responses. Using mouse and monkey models, we found that the activation of both CD4 and CD8 T cells was amplified by combination with anti-CTLA-4 and anti-PD-1 antibodies. These antibodies were administered subcutaneously to target their distribution to vaccination sites and to reduce systemic exposure which may improve their safety. VBIR can break tolerance and activate T cells recognizing tumor-associated self-antigens. This activation lasts more than a year after completing treatment in monkeys, and inhibits tumor growth to a greater degree than is observed using the individual components in mouse cancer models. These results have encouraged the testing of this combination regimen in cancer patients with the aim of increasing responses beyond current therapies.


Assuntos
Vacinas Anticâncer , Neoplasias , Vacinas de DNA , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Antígenos de Neoplasias , Vacinação/métodos , Modelos Animais de Doenças , Autoantígenos
2.
Trends Immunol ; 39(7): 536-548, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29751996

RESUMO

Antitumor rejection by the immune system is a complex process that is regulated by several factors. Among these factors are the quality and quantity of mutational events that occur in cancer cells. Perhaps one of the most important types of mutations that influence antitumor immunity is the neoantigen, that is, a non-self-antigen that arises as a result of somatic mutation. Recent work has demonstrated that neoantigens hold significant promise for developing new diagnostic and therapeutic modalities. Therapeutic targeting of neoantigens is important for achieving benefit following therapy with immune checkpoint blockade agents or for cancer vaccines targeting mutations. Here, we review our understanding of neoantigens and discuss new developments in the quest to use them in cancer immunotherapy.


Assuntos
Antineoplásicos/imunologia , Vacinas Anticâncer/imunologia , Neoplasias/imunologia , Animais , Antígenos de Neoplasias/imunologia , Humanos , Imunoterapia/métodos , Neoplasias/terapia
3.
Nat Med ; 30(4): 1013-1022, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38538867

RESUMO

Therapeutic vaccines that elicit cytotoxic T cell responses targeting tumor-specific neoantigens hold promise for providing long-term clinical benefit to patients with cancer. Here we evaluated safety and tolerability of a therapeutic vaccine encoding 20 shared neoantigens derived from selected common oncogenic driver mutations as primary endpoints in an ongoing phase 1/2 study in patients with advanced/metastatic solid tumors. Secondary endpoints included immunogenicity, overall response rate, progression-free survival and overall survival. Eligible patients were selected if their tumors expressed one of the human leukocyte antigen-matched tumor mutations included in the vaccine, with the majority of patients (18/19) harboring a mutation in KRAS. The vaccine regimen, consisting of a chimp adenovirus (ChAd68) and self-amplifying mRNA (samRNA) in combination with the immune checkpoint inhibitors ipilimumab and nivolumab, was shown to be well tolerated, with observed treatment-related adverse events consistent with acute inflammation expected with viral vector-based vaccines and immune checkpoint blockade, the majority grade 1/2. Two patients experienced grade 3/4 serious treatment-related adverse events that were also dose-limiting toxicities. The overall response rate was 0%, and median progression-free survival and overall survival were 1.9 months and 7.9 months, respectively. T cell responses were biased toward human leukocyte antigen-matched TP53 neoantigens encoded in the vaccine relative to KRAS neoantigens expressed by the patients' tumors, indicating a previously unknown hierarchy of neoantigen immunodominance that may impact the therapeutic efficacy of multiepitope shared neoantigen vaccines. These data led to the development of an optimized vaccine exclusively targeting KRAS-derived neoantigens that is being evaluated in a subset of patients in phase 2 of the clinical study. ClinicalTrials.gov registration: NCT03953235 .


Assuntos
Vacinas Anticâncer , Neoplasias , Vacinas , Humanos , Antígenos de Neoplasias , Vacinas Anticâncer/efeitos adversos , Antígenos HLA , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Vacinas/uso terapêutico
4.
Clin Immunol ; 148(2): 287-98, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23811319

RESUMO

In this report, a Treg-depleting anti-FR4 antibody is combined with a GM-CSF-secreting tumor cell immunotherapy (GVAX) for treatment of melanoma-bearing animals. Median survival time (MST) of animals treated with GVAX was 41 days, compared to a MST of 32 days in untreated animals. Anti-FR4 monotherapy had no effect on MST. Combination of anti-FR4 and GVAX significantly prolonged MST to 55 days, suggesting that these two agents can function cooperatively. Combination therapy increased expression of IFN-γ and granzyme B by CD8 T cells. In contrast to anti-CD25-mediated Treg depletion, administration of anti-FR4 after GVAX did not reduce efficacy, suggesting that anti-FR4 does not deplete effector cells induced by GVAX. Triple combination of a blocking CTLA4 antibody with GVAX and anti-FR4 further enhanced overall survival and reduced growth of well-established melanomas. Considered together, anti-FR4 antibody and GVAX may be a promising approach for the treatment of patients with cancer.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunoterapia/métodos , Neoplasias/terapia , Receptores de Superfície Celular/metabolismo , Linfócitos T Reguladores/fisiologia , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia
5.
Cancer Immunol Immunother ; 62(2): 245-56, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22878899

RESUMO

Immune checkpoint blockade enhances antitumor responses, but can also lead to severe immune-related adverse events (IRAE). To avoid unnecessary exposure to these potentially hazardous agents, it is important to identify biomarkers that correlate with clinical activity and can be used to select patients that will benefit from immune checkpoint blockade. To understand the consequences of CTLA-4 blockade and identify biomarkers for clinical efficacy and/or survival, an exploratory T cell monitoring study was performed in a phase I/II dose escalation/expansion trial (n = 28) of combined Prostate GVAX/ipilimumab immunotherapy. Phenotypic T cell monitoring in peripheral blood before and after Prostate GVAX/ipilimumab treatment revealed striking differences between patients who benefited from therapy and patients that did not. Treatment-induced rises in absolute lymphocyte counts, CD4(+) T cell differentiation, and CD4(+) and CD8(+) T cell activation were all associated with clinical benefit. Moreover, significantly prolonged overall survival (OS) was observed for patients with high pre-treatment frequencies of CD4(+)CTLA-4(+), CD4(+)PD-1(+), or differentiated (i.e., non-naive) CD8(+) T cells or low pre-treatment frequencies of differentiated CD4(+) or regulatory T cells. Unsupervised clustering of these immune biomarkers revealed cancer-related expression of CTLA-4(+) in CD4(+) T cells to be a dominant predictor for survival after Prostate GVAX/ipilimumab therapy and to thus provide a putative and much-needed biomarker for patient selection prior to therapeutic CTLA4 blockade.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Linfócitos T CD4-Positivos/efeitos dos fármacos , Antígeno CTLA-4/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias da Próstata/terapia , Idoso , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/análise , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Ipilimumab , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/mortalidade
6.
Lancet Oncol ; 13(5): 509-17, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22326922

RESUMO

BACKGROUND: The granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells vaccine (GVAX) has antitumour activity against prostate cancer; preclinical studies have shown potent synergy when combined with ipilimumab, an antibody that blocks cytotoxic T-lymphocyte antigen 4. We aimed to assess the safety of combined treatment with GVAX and ipilimumab in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS: We did an open-labelled, single-centre, dose-escalation study of ipilimumab concurrent with a fixed dose of GVAX, with a subsequent expansion phase, both at the VU University Medical Centre (Amsterdam, Netherlands). Eligible patients had documented mCRPC and had not been previously treated with chemotherapy. All patients received a 5×10(8) cell priming dose of GVAX intradermally on day 1 with subsequent intradermal injections of 3×10(8) cells every 2 weeks for 24 weeks. The vaccinations were combined with intravenous ipilimumab every 4 weeks. We enrolled patients in cohorts of three; each cohort received an escalating dose of ipilimumab at 0·3, 1·0, 3·0, or 5·0 mg/kg. Our primary endpoint was safety. This study is registered with ClinicalTrials.gov, number NCT01510288. FINDINGS: We enrolled 12 patients into our dose-escalation cohort. We did not record any severe immune-related adverse events at the first two dose levels. At the 3·0 mg/kg dose level, one patient had grade 2 and two patients grade 3 hypophysitis; at the 5·0 mg/kg dose level, two patients had grade 3 hypophysitis and one patient developed grade 4 sarcoid alveolitis (a dose-limiting toxic effect). Due to observed clinical activity and toxic events, we decided to expand the 3·0 mg/kg dose level, rather than enrol a further three patients at the 5·0 mg/kg level. 16 patients were enrolled in the expansion cohort, two of whom developed grade 2 hypophysitis, three colitis (one grade 1 and two grade 2), and one grade 3 hepatitis--all immune-related adverse events. The most common adverse events noted in all 28 patients were injection-site reactions (grade 1-2 events seen in all patients), fatigue (grade 1-2 in 20 patients, grade 3 in two), and pyrexia (grade 1-2 in 15 patients, grade 3 in one). 50% or greater declines in prostate-specific antigen from baseline was recorded in seven patients (25%); all had received 3·0 mg/kg or 5·0 mg/kg ipilimumab. INTERPRETATION: GVAX combined with 3·0 mg/kg ipilimumab is tolerable and safe for patients with mCRPC. Further research on the combined treatment of patients with mCRPC with vaccination and ipilimumab is warranted. FUNDING: Cell Genesys Inc, Prostate Cancer Foundation, Dutch Cancer Society (KWF-VU 2006-3697), and Foundation Stichting VUmc Cancer Center Amsterdam.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias da Próstata/terapia , Adulto , Idoso , Terapia Combinada , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunoterapia , Ipilimumab , Masculino , Pessoa de Meia-Idade , Orquiectomia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/secundário , Transplante Homólogo , Células Tumorais Cultivadas
7.
Nat Commun ; 14(1): 3274, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280238

RESUMO

SARS-CoV-2 has resulted in high levels of morbidity and mortality world-wide, and severe complications can occur in older populations. Humoral immunity induced by authorized vaccines wanes within 6 months, and frequent boosts may only offer transient protection. GRT-R910 is an investigational self-amplifying mRNA (samRNA)-based SARS-CoV-2 vaccine delivering full-length Spike and selected conserved non-Spike T cell epitopes. This study reports interim analyses for a phase I open-label dose-escalation trial evaluating GRT-R910 in previously vaccinated healthy older adults (NCT05148962). Primary endpoints of safety and tolerability were assessed. Most solicited local and systemic adverse events (AEs) following GRT-R910 dosing were mild to moderate and transient, and no treatment-related serious AEs were observed. The secondary endpoint of immunogenicity was assessed via IgG binding assays, neutralization assays, interferon-gamma ELISpot, and intracellular cytokine staining. Neutralizing antibody titers against ancestral Spike and variants of concern were boosted or induced by GRT-R910 and, contrasting to authorized vaccines, persisted through at least 6 months after the booster dose. GRT-R910 increased and/or broadened functional Spike-specific T cell responses and primed functional T cell responses to conserved non-Spike epitopes. This study is limited due to small sample size, and additional data from ongoing studies will be required to corroborate these interim findings.


Assuntos
COVID-19 , RNA Mensageiro/genética , COVID-19/prevenção & controle , Humanos , Idoso , Masculino , Feminino , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Ensaios Clínicos como Assunto , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Linfócitos T/imunologia
8.
Nat Commun ; 13(1): 3289, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672369

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self-amplifying mRNA (SAM) vaccine encoding a prefusion stabilized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein and demonstrate strong cellular and humoral immune responses at low doses in mice and rhesus macaques. The homologous prime-boost vaccination regimen of SAM at 3, 10 and 30 µg induced potent neutralizing antibody (nAb) titers in rhesus macaques following two SAM vaccinations at all dose levels, with the 10 µg dose generating geometric mean titers (GMT) 48-fold greater than the GMT of a panel of SARS-CoV-2 convalescent human sera. Spike-specific T cell responses were observed with all tested vaccine regimens. SAM vaccination provided protective efficacy against SARS-CoV-2 challenge as both a homologous prime-boost and as a single boost following ChAd prime, demonstrating reduction of viral replication in both the upper and lower airways. The SAM vaccine is currently being evaluated in clinical trials as both a homologous prime-boost regimen at low doses and as a boost following heterologous prime.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Humanos , Macaca mulatta/genética , Camundongos , RNA Mensageiro , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Vacinação
9.
Sci Immunol ; 7(73): eabn8097, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35857579

RESUMO

Preclinical studies show that locoregional CTLA-4 blockade is equally effective in inducing tumor eradication as systemic delivery, without the added risk of immune-related side effects. This efficacy is related to access of the CTLA-4 blocking antibodies to tumor-draining lymph nodes (TDLNs). Local delivery of anti-CTLA-4 after surgical removal of primary melanoma, before sentinel lymph node biopsy (SLNB), provides a unique setting to clinically assess the role of TDLN in the biological efficacy of locoregional CTLA-4 blockade. Here, we have evaluated the safety, tolerability, and immunomodulatory effects in the SLN and peripheral blood of a single dose of tremelimumab [a fully human immunoglobulin gamma-2 (IgG2) mAb directed against CTLA-4] in a dose range of 2 to 20 mg, injected intradermally at the tumor excision site 1 week before SLNB in 13 patients with early-stage melanoma (phase 1 trial; NCT04274816). Intradermal delivery was safe and well tolerated and induced activation of migratory dendritic cell (DC) subsets in the SLN. It also induced profound and durable decreases in regulatory T cell (Treg) frequencies and activation of effector T cells in both SLN and peripheral blood. Moreover, systemic T cell responses against NY-ESO-1 or MART-1 were primed or boosted (N = 7), in association with T cell activation and central memory T cell differentiation. These findings indicate that local administration of anti-CTLA-4 may offer a safe and promising adjuvant treatment strategy for patients with early-stage melanoma. Moreover, our data demonstrate a central role for TDLN in the biological efficacy of CTLA-4 blockade and support TDLN-targeted delivery methods.


Assuntos
Imunoterapia , Linfonodos , Melanoma , Anticorpos Monoclonais Humanizados/administração & dosagem , Humanos , Imunoterapia/métodos , Injeções Intradérmicas/efeitos adversos , Linfonodos/patologia , Ativação Linfocitária , Melanoma/patologia , Melanoma/terapia , Biópsia de Linfonodo Sentinela
10.
Nat Med ; 28(8): 1619-1629, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35970920

RESUMO

Checkpoint inhibitor (CPI) therapies provide limited benefit to patients with tumors of low immune reactivity. T cell-inducing vaccines hold promise to exert long-lasting disease control in combination with CPI therapy. Safety, tolerability and recommended phase 2 dose (RP2D) of an individualized, heterologous chimpanzee adenovirus (ChAd68) and self-amplifying mRNA (samRNA)-based neoantigen vaccine in combination with nivolumab and ipilimumab were assessed as primary endpoints in an ongoing phase 1/2 study in patients with advanced metastatic solid tumors (NCT03639714). The individualized vaccine regimen was safe and well tolerated, with no dose-limiting toxicities. Treatment-related adverse events (TRAEs) >10% included pyrexia, fatigue, musculoskeletal and injection site pain and diarrhea. Serious TRAEs included one count each of pyrexia, duodenitis, increased transaminases and hyperthyroidism. The RP2D was 1012 viral particles (VP) ChAd68 and 30 µg samRNA. Secondary endpoints included immunogenicity, feasibility of manufacturing and overall survival (OS). Vaccine manufacturing was feasible, with vaccination inducing long-lasting neoantigen-specific CD8 T cell responses. Several patients with microsatellite-stable colorectal cancer (MSS-CRC) had improved OS. Exploratory biomarker analyses showed decreased circulating tumor DNA (ctDNA) in patients with prolonged OS. Although small study size limits statistical and translational analyses, the increased OS observed in MSS-CRC warrants further exploration in larger randomized studies.


Assuntos
Neoplasias Colorretais , Pan troglodytes , Adenoviridae/genética , Animais , Neoplasias Colorretais/tratamento farmacológico , Febre , Humanos , RNA Mensageiro/uso terapêutico
11.
Cancer Immunol Immunother ; 59(9): 1313-23, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20499060

RESUMO

A critical factor in clinical development of cancer immunotherapies is the identification of tumor-associated antigens that may be related to immunotherapy potency. In this study, protein microarrays containing >8,000 human proteins were screened with serum from prostate cancer patients (N = 13) before and after treatment with a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting whole cell immunotherapy. Thirty-three proteins were identified that displayed significantly elevated (P

Assuntos
Anticorpos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Proteínas de Choque Térmico HSP90/biossíntese , Imunoterapia Adotiva , Proteínas Nucleares/biossíntese , Neoplasias da Próstata/imunologia , Anticorpos/imunologia , Biomarcadores Farmacológicos , Biomarcadores Tumorais/imunologia , Ensaios Clínicos como Assunto , Progressão da Doença , Galectinas/imunologia , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/imunologia , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Metástase Neoplásica , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Prognóstico , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Análise Serial de Proteínas , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
12.
J Virol ; 83(8): 3450-62, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19193803

RESUMO

Oncolytic adenoviral vectors that express immunostimulatory transgenes are currently being evaluated in clinic. Preclinical testing of these vectors has thus far been limited to immunodeficient xenograft tumor models since human adenoviruses do not replicate effectively in murine tumor cells. The effect of the immunostimulatory transgene on overall virus potency can therefore not be readily assessed in these models. Here, a model is described that allows the effective testing of mouse armed oncolytic adenovirus (MAV) vectors in immunocompetent syngeneic tumor models. These studies demonstrate that the MAV vectors have a high level of cytotoxicity in a wide range of murine tumor cells. The murine oncolytic viruses were successfully armed with murine granulocyte-macrophage colony-stimulating factor (mGM-CSF) by a novel method which resulted in vectors with a high level of tumor-specific transgene expression. The mGM-CSF-armed MAV vectors showed an improved level of antitumor potency and induced a systemic antitumor immune response that was greater than that induced by unarmed parental vectors in immunocompetent syngeneic tumor models. Thus, the oncolytic MAV-1 system described here provides a murine homolog model for the testing of murine armed oncolytic adenovirus vectors in immunocompetent animals. The model allows evaluation of the impact of virus replication and the host immune response on overall virus potency and enables the generation of translational data that will be important for guiding the clinical development of these viruses.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Modelos Animais , Neoplasias/virologia , Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/imunologia , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Vírus Oncolíticos/genética
13.
Clin Cancer Res ; 15(5): 1623-34, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19208793

RESUMO

PURPOSE: The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy, which is known to stimulate potent and long-lasting antigen-specific immune responses, in combination with PD-1 blockade, which has been shown to augment cellular immune responses. EXPERIMENTAL DESIGN: Survival studies were done in the B16 melanoma and CT26 colon carcinoma tumor models. Immune monitoring studies were done in the B16 model. GM-CSF-secreting tumor cell immunotherapy was administered s.c. and the anti-PD-1 antibody was administered i.p. RESULTS: The studies reported here show that combining PD-1 blockade with GM-CSF-secreting tumor cell immunotherapy prolonged the survival of tumor-bearing animals compared with animals treated with either therapy alone. Prolonged survival correlated with strong antigen-specific T-cell responses detected by tetramer staining and an in vivo CTL assay, higher secretion levels of proinflammatory cytokines by splenocytes, and the persistence of functional CD8+ T cells in the tumor microenvironment. Furthermore, in the biweekly multiple treatment setting, repeated antigen-specific T-cell expansion was only observed following administration of the cellular immunotherapy with the PD-1 blockade and not when the cellular immunotherapy or PD-1 blockade was used as monotherapy. CONCLUSION: The combination of PD-1 blockade with GM-CSF-secreting tumor cell immunotherapy leads to significantly improved antitumor responses by augmenting the tumor-reactive T-cell responses induced by the cellular immunotherapy. Readministration of the cellular immunotherapy with the anti-PD-1 antibody in subsequent immunotherapy cycles was required to reactivate these T-cell responses.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Neoplasias do Colo/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Imunoterapia , Melanoma Experimental/terapia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Proteínas Reguladoras de Apoptose/imunologia , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Citocinas/metabolismo , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo , Receptor de Morte Celular Programada 1 , Ratos , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Citotóxicos/imunologia
14.
Appl Microbiol Biotechnol ; 87(4): 1517-24, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20461511

RESUMO

The recently described 2A/furin technology combines both chains of the antibody in a single open reading frame. Upon translation and secretion, the peptide is processed by the cell to generate native fully functional IgG antibodies. Here, we describe the results of an evaluation study of this technology for an industrial CHO cell line development process. The 2A/furin expression cassette setup was combined with a Novartis vector system. A transfection, selection, and cloning procedure in chemically defined media was established at Novartis and applied for a monoclonal test antibody. The productivity of 2A/furin-vector-derived clones in non-optimized generic shake flask fed-batch models was in a comparable range with clones derived from the reference control vector. Higher clonal production stability was seen for the majority of clones generated with the 2A/furin technology compared to the clones generated with the reference control vector. Product quality was analyzed by SDS-PAGE and no significant difference was detected between the two systems. Thus, it was shown that the 2A/furin technology can be successfully combined with a Novartis CHO expression system and platform. Due to the single ORF setup, the 2A/furin technology may therefore offer a suitable approach to reduce vector size and complexity.


Assuntos
Biotecnologia/métodos , Furina/genética , Vetores Genéticos/genética , Animais , Células CHO , Cricetinae , Cricetulus , Furina/metabolismo , Expressão Gênica , Vetores Genéticos/metabolismo
15.
Clin Immunol ; 133(2): 184-97, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19664962

RESUMO

Clinical studies of cell-based immunotherapies have included both patient-specific (autologous) and non-patient-specific (allogeneic) approaches. Major concerns in using allogeneic immunotherapies are that the induced immune responses may be predominantly directed against the allogeneic HLA molecules of the cellular immunotherapy and not against its potential tumor antigens and that only the allogeneic responses will be enhanced when the immunotherapies are combined with immune checkpoint regulators in an effort to enhance overall immunotherapy potency. To evaluate these possibilities, studies were performed using the GM-CSF-secreting B16F1 cell line as autologous immunotherapy (Auto) and the same cell line modified to over-express the MHC molecule K(d) to generate an immunotherapy that expresses an allogeneic component (Allo) when injected into C57/Bl6 mice. The goal was to compare the specific anti-tumor immune responses induced by these two immunotherapies, which share an identical antigen repertoire, with the exception of the allogeneic MHC class I molecule expressed by the Allo cells, and have identical GM-CSF-secretion levels. Both immunotherapies provided similar therapeutic benefit to tumor-bearing animals with a trend towards a more pronounced tumor growth delay in animals injected with the Allo immunotherapy. This correlated with a significant increase in the number of activated DCs and T-cells in the DLN of Allo-treated animals. In addition, persistent infiltration of effector CD8(+) T-cells was detected in the tumors of animals treated with the Allo immunotherapy, which correlated with a trend towards a greater antigen-specific T-cell response in these animals. When combined with the immune checkpoint regulator anti-PD-1, tumor-specific and allogeneic immune responses were equally enhanced. Thus, the ability of an allogeneic tumor cell immunotherapy to induce a therapeutic anti-tumor immune response is comparable, if not superior, to an autologous tumor cell immunotherapy and its anti-tumor potency can be enhanced when combined with immunomodulatory compounds.


Assuntos
Vacinas Anticâncer/imunologia , Transplante de Células/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Transplante Autólogo/imunologia , Transplante Homólogo/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/imunologia , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoterapia/métodos , Linfonodos/citologia , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Baço/citologia , Baço/imunologia , Análise de Sobrevida , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Transfecção
16.
Clin Cancer Res ; 14(11): 3545-54, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18519788

RESUMO

PURPOSE: The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy, which is known to stimulate a potent and long-lasting antigen-specific immune response in combination with lymphocyte activation gene-3 fusion protein (LAG-3Ig), which has been shown to act as an adjuvant for priming T helper type 1 and cytotoxic T-cell responses. EXPERIMENTAL DESIGN: Survival and immune monitoring studies were done in the B16 melanoma model. GM-CSF-secreting tumor cell immunotherapy was administered as a single s.c. injection and LAG-3Ig was administered s.c. at the immunotherapy site. RESULTS: The studies reported here show that combining LAG-3Ig with GM-CSF-secreting tumor cell immunotherapy prolonged the survival of tumor-bearing animals compared with animals treated with either therapy alone. Prolonged survival correlated with increased numbers of systemic IFN gamma-secreting CD8+ T cells and a significantly increased infiltration of activated effector CD8+ T cells into the tumor. Moreover, an increase in antigen-specific IgG1 humoral responses was detected in serum of animals injected with the combination therapy compared with animals injected with either therapy alone. CONCLUSION: LAG-3Ig combined with a GM-CSF-secreting tumor cell immunotherapy stimulated both cellular and humoral antitumor immune responses that correlated with prolonged survival in tumor-bearing animals.


Assuntos
Antígenos CD/uso terapêutico , Imunoterapia/métodos , Fator Estimulador de Colônias de Macrófagos/uso terapêutico , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Anticorpos/sangue , Terapia Combinada , Citocinas/biossíntese , Citotoxicidade Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
17.
Clin Cancer Res ; 14(12): 3933-41, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559615

RESUMO

PURPOSE: Evaluate the codelivery of hyaluronidase enzyme with oncolytic adenoviruses to determine whether it improves the spread of the virus throughout tumors, thereby leading to a greater overall antitumor efficacy in tumor models. EXPERIMENTAL DESIGN: The optimal dose of hyaluronidase that provided best transduction efficiency and spread of a green fluorescent protein (GFP)-expressing adenovirus within tumors was combined with oncolytic viruses in tumor models to determine whether the combination treatment results in an improvement of antitumor efficacy. RESULTS: In mice injected with the adenovirus Ad5/35GFP and an optimal dose of hyaluronidase (50 U), a significant increase in the number of GFP-expressing cells was observed when compared with animals injected with virus only (P < 0.0001). When the oncolytic adenoviruses Ad5OV or Ad5/35 OV (OV-5 or OV5T35H) were codelivered with 50 U of hyaluronidase, a significant delay in tumor progression was observed, which translated into a significant increase in the mean survival time of tumor-bearing mice compared with either of the monotherapy-treated groups (P < 0.0001). Furthermore, the mice that received the combination of Ad5/35 OV and hyaluronidase showed the best antitumor efficacy. Importantly, the combination treatment did not increase the metastatic potential of the tumors. Lastly, the increase in virus potency observed in animals injected with both enzyme and virus correlated with enhanced virus spread throughout tumors. CONCLUSION: Antitumor activity and overall survival of mice bearing highly aggressive tumors are significantly improved by codelivery of oncolytic adenoviruses and hyaluronidase when compared with either of the monotherapy-treated groups, and it may prove to be a potent and novel approach to treating patients with cancer.


Assuntos
Adenoviridae/efeitos dos fármacos , Hialuronoglucosaminidase/administração & dosagem , Hialuronoglucosaminidase/farmacologia , Vírus Oncolíticos/efeitos dos fármacos , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Adenoviridae/fisiologia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Injeções Intralesionais , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Resultado do Tratamento , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Res ; 67(9): 4399-407, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483354

RESUMO

Selective replication of oncolytic viruses in tumor cells provides a promising approach for the treatment of human cancers. One of the limitations observed with oncolytic viruses currently used in the treatment of solid tumors is the inefficient spread of virus throughout the tumor mass following intratumoral injection. Data are presented showing that oncolytic adenoviruses expressing the relaxin gene and containing an Ad5/Ad35 chimeric fiber showed significantly enhanced transduction and increased virus spread throughout the tumor when compared with non-relaxin-expressing, Ad5-based viruses. The increased spread of such viruses throughout tumors correlated well with improved antitumor efficacy and overall survival in two highly metastatic tumor models. Furthermore, nonreplicating viruses expressing relaxin did not increase metastases, suggesting that high level expression of relaxin will not enhance metastatic spread of tumors. In summary, the data show that relaxin may play a role in rearranging matrix components within tumors, which helps recombinant oncolytic adenoviruses to spread effectively throughout the tumor mass and thereby increase the extent of viral replication within the tumor. Expressing relaxin from Ad5/Ad35 fiber chimeric adenoviruses may prove a potent and novel approach to treating patients with cancer.


Assuntos
Adenoviridae/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Relaxina/biossíntese , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Linhagem Celular Tumoral , Colágeno/metabolismo , Feminino , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/virologia , Relaxina/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Exp Hematol ; 36(3): 319-28, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18279719

RESUMO

OBJECTIVE: Acute myeloid leukemia (AML) is a highly malignant neoplasm responsible for nearly 10,000 cancer-related deaths annually in the United States. Treatment options for elderly patients with AML remain limited. Standard regimens using cytarabine (cytosine arabinoside [AraC]), a nucleotide analogue, result in significant toxicity with poor overall response. Combination of a cytotoxic chemotherapy and tumor-specific immunotherapy has the potential to improve overall efficacy by inducing an anti-tumor immune response against minimal residual disease. The studies reported here were performed to evaluate the therapeutic benefit of combining a granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy with AraC treatment. MATERIALS AND METHODS: C57Bl/6 mice were challenged with C1498-luc cells intravenously and evaluated by in vivo imaging throughout the study to monitor the systemic progression of the tumor. Individual animals were euthanized when in vivo total photon counts exceeded 5 x 10(8) and/or when they were in poor clinical condition. Cytotoxicity assay was performed to evaluate effector function and flow cytometry was used for phenotyping of splenocytes from experimental animals. RESULTS: Administration of GM-CSF-secreting tumor cell immunotherapy during AraC -induced cytopenia enhanced the anti-tumor efficacy of the immunotherapy, resulting in prolonged survival. AraC treatment did not negatively impact antigen-specific T-cell activation elicited by the immunotherapy and surviving animals treated with the combination demonstrated strong tumor-specific memory responses. CONCLUSION: GM-CSF-secreting tumor cell immunotherapy in combination with AraC prolongs survival of tumor-bearing mice, with a median survival time of 61 days observed in mice treated with AraC alone and 90% of mice treated with the combination therapy still alive by day 150.


Assuntos
Antineoplásicos/administração & dosagem , Citarabina/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Imunoterapia/métodos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Animais , Antineoplásicos/efeitos adversos , Linhagem Celular Tumoral , Terapia Combinada , Citarabina/efeitos adversos , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Memória Imunológica , Injeções Intraperitoneais , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Luciferases/biossíntese , Luciferases/genética , Linfopenia/induzido quimicamente , Linfopenia/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fenótipo , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento
20.
Nat Biotechnol ; 23(5): 584-90, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15834403

RESUMO

Therapeutic monoclonal antibodies (mAbs) are currently being developed for the treatment of cancer and other diseases. Despite clinical success, widespread application of mAb therapies may be limited by manufacturing capabilities. In this paper, we describe a mAb delivery system that allows continuous production of a full-length antibody at high-concentrations in vivo after gene transfer. The mAb is expressed from a single open reading frame by linking the heavy and light chains with a 2A self-processing peptide derived from the foot-and-mouth disease virus. Using this expression system, we generated a recombinant adeno-associated virus vector encoding the VEGFR2-neutralizing mAb DC101 (rAAV8-DC101). A single dose of rAAV8-DC101 resulted in long-term expression of >1,000 microg/ml of DC101 in mice, demonstrating significant anti-tumor efficacy. This report describes the first feasible gene therapy approach for stable delivery of mAbs at therapeutic levels, which may serve as an attractive alternative to direct injection of mAbs.


Assuntos
Adenoviridae/genética , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/uso terapêutico , Formação de Anticorpos/genética , Terapia Genética/métodos , Rim/metabolismo , Neoplasias/terapia , Proteínas Virais/metabolismo , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/isolamento & purificação , Linhagem Celular , Técnicas de Transferência de Genes , Humanos , Hibridomas , Rim/imunologia , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa