Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
1.
Curr Opin Immunol ; 84: 102376, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37598549

RÉSUMÉ

Delivery of vaccines via the mucosal route is regarded as the most effective mode of immunization to counteract infectious diseases that enter via mucosal tissues, including oral, nasal, pulmonary, intestinal, and urogenital surfaces. Mucosal vaccines not only induce local immune effector elements, such as secretory Immunoglobulin A (IgA) reaching the luminal site of the mucosa, but also systemic immunity. Moreover, mucosal vaccines may trigger immunity in distant mucosal tissues because of the homing of primed antigen-specific immune cells toward local and distant mucosal tissue via the common mucosal immune system. While most licensed intramuscular vaccines induce only systemic immunity, next-generation mucosal vaccines may outperform parenteral vaccination strategies by also eliciting protective mucosal immune responses that block infection and/or transmission. Especially the nasal route of vaccination, targeting the nasal-associated lymphoid tissue, is attractive for local and distant mucosal immunization. In numerous studies, bacterial outer membrane vesicles (OMVs) have proved attractive as vaccine platform for homologous bacterial strains, but also as antigen delivery platform for heterologous antigens of nonbacterial diseases, including viruses, parasites, and cancer. Their application has also been extended to mucosal delivery. Here, we will summarize the characteristics and clinical potential of (engineered) OMVs as vaccine platform for mucosal, especially intranasal delivery.


Sujet(s)
Vaccins , Humains , Administration par voie nasale , Immunisation , Vaccination , Immunité muqueuse , Muqueuse
2.
Front Oncol ; 12: 934638, 2022.
Article de Anglais | MEDLINE | ID: mdl-35837107

RÉSUMÉ

Background: Glioblastoma (GBM) is the most common primary, malignant brain tumor in adults and has a poor prognosis. The median progression-free survival (mPFS) of newly diagnosed GBM is approximately 6 months. The recurrence rate approaches 100%, and the case-fatality ratio approaches one. Half the patients die within 8 months of recurrence, and 5-year survival is less than 10%. Advances in treatment options are urgently needed. We report on the efficacy and safety of a therapeutic vaccine (SITOIGANAP: Epitopoietic Research Corporation) administered to 21 patients with recurrent GBM (rGBM) under a Right-to-Try/Expanded Access program. SITOIGANAP is composed of both autologous and allogeneic tumor cells and lysates. Methods: Twenty-one patients with rGBM received SITOIGANAP on 28-day cycles in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), cyclophosphamide, bevacizumab, and an anti-programmed cell death protein-1 (anti-PD-1) monoclonal antibody (either nivolumab or pembrolizumab). Results: The mPFS was 9.14 months, and the median overall survival (mOS) was 19.63 months from protocol entry. Currently, 14 patients (67%) are at least 6 months past their first SITOIGANAP cycle; 10 patients (48%) have received at least six cycles and have a mOS of 30.64 months and 1-year survival of 90%. The enrollment and end-of-study CD3+/CD4+ T-lymphocyte counts strongly correlate with OS. Conclusions: The addition of SITOIGANAP/GM-CSF/cyclophosphamide to bevacizumab and an anti-PD-1 monoclonal antibody resulted in a significant survival benefit compared to historic control values in rGBM with minimal toxicity compared to current therapy.

3.
Pharmaceutics ; 13(4)2021 Apr 06.
Article de Anglais | MEDLINE | ID: mdl-33917629

RÉSUMÉ

Vaccines are the most effective medical intervention due to their continual success in preventing infections and improving mortality worldwide. Early vaccines were developed empirically however, rational design of vaccines can allow us to optimise their efficacy, by tailoring the immune response. Establishing the immune correlates of protection greatly informs the rational design of vaccines. This facilitates the selection of the best vaccine antigens and the most appropriate vaccine adjuvant to generate optimal memory immune T cell and B cell responses. This review outlines the range of vaccine types that are currently authorised and those under development. We outline the optimal immunological correlates of protection that can be targeted. Finally we review approaches to rational antigen selection and rational vaccine adjuvant design. Harnessing current knowledge on protective immune responses in combination with critical vaccine components is imperative to the prevention of future life-threatening diseases.

4.
Immunol Rev ; 296(1): 169-190, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32594569

RÉSUMÉ

Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.


Sujet(s)
Adjuvants immunologiques , Immunomodulation , Vaccination , Vaccins/immunologie , Vaccins/usage thérapeutique , Animaux , Production d'anticorps/immunologie , Auto-immunité , Prise en charge de la maladie , Humains , Immunité cellulaire , Immunité humorale , Thérapie moléculaire ciblée , Résultat thérapeutique , Vaccination/méthodes , Vaccins/administration et posologie
5.
Eur J Immunol ; 50(7): 932-938, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32438473

RÉSUMÉ

The recent outbreak of coronavirus disease 2019 (COVID-19), triggered by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses an enormous threat to global public health and economies. Human coronaviruses normally cause no or mild respiratory disease but in the past two decades, potentially fatal coronavirus infections have emerged, causing respiratory tract illnesses such as pneumonia and bronchitis. These include severe acute respiratory syndrome coronavirus (SARS-CoV), followed by the Middle East respiratory syndrome coronavirus (MERS-CoV), and recently the SARS-CoV-2 coronavirus outbreak that emerged in Wuhan, China, in December 2019. Currently, most COVID-19 patients receive traditional supportive care including breathing assistance. To halt the ongoing spread of the pandemic SARS-CoV-2 coronavirus and rescue individual patients, established drugs and new therapies are under evaluation. Since it will be some time until a safe and effective vaccine will be available, the immediate priority is to harness innate immunity to accelerate early antiviral immune responses. Second, since excessive inflammation is a major cause of pathology, targeted anti-inflammatory responses are being evaluated to reduce inflammation-induced damage to the respiratory tract and cytokine storms. Here, we highlight prominent immunotherapies at various stages of development that aim for augmented anti-coronavirus immunity and reduction of pathological inflammation.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Antiviraux/usage thérapeutique , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/immunologie , Immunité innée/effets des médicaments et des substances chimiques , Immunothérapie/méthodes , Pneumopathie virale/traitement médicamenteux , Pneumopathie virale/immunologie , Animaux , Anti-inflammatoires/effets indésirables , Antiviraux/effets indésirables , Betacoronavirus/physiologie , COVID-19 , Infections à coronavirus/anatomopathologie , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/thérapie , Humains , Immunisation passive , Immunomodulation , Pandémies/prévention et contrôle , Pneumopathie virale/anatomopathologie , Pneumopathie virale/prévention et contrôle , SARS-CoV-2 , Sérothérapie COVID-19
6.
Int J Mol Sci ; 19(9)2018 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-30150597

RÉSUMÉ

Glioblastoma is the most common form of brain cancer in adults that produces severe damage to the brain leading to a very poor survival prognosis. The standard of care for glioblastoma is usually surgery, as well as radiotherapy followed by systemic temozolomide chemotherapy, resulting in a median survival time of about 12 to 15 months. Despite these therapeutic efforts, the tumor returns in the vast majority of patients. When relapsing, statistics suggest an imminent death dependent on the size of the tumor, the Karnofsky Performance Status, and the tumor localization. Following the standard of care, the administration of Bevacizumab, inhibiting the growth of the tumor vasculature, is an approved medicinal treatment option approved in the United States, but not in the European Union, as well as the recently approved alternating electric fields (AEFs) generator NovoTTF/Optune. However, it is clear that regardless of the current treatment regimens, glioma patients continue to have dismal prognosis and novel treatments are urgently needed. Here, we describe different approaches of recently developed therapeutic glioma brain cancer vaccines, which stimulate the patient's immune system to recognize tumor-associated antigens (TAA) on cancer cells, aiming to instruct the immune system to eventually attack and destroy the brain tumor cells, with minimal bystander damage to normal brain cells. These distinct immunotherapies may target particular glioma TAAs which are molecularly defined, but they may also target broad patient-derived tumor antigen preparations intentionally evoking a very broad polyclonal antitumor immune stimulation.


Sujet(s)
Tumeurs du cerveau/immunologie , Vaccins anticancéreux/immunologie , Glioblastome/immunologie , Immunisation/méthodes , Encéphale/effets des médicaments et des substances chimiques , Encéphale/immunologie , Encéphale/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Vaccins anticancéreux/usage thérapeutique , Glioblastome/traitement médicamenteux , Humains , Système immunitaire/effets des médicaments et des substances chimiques , Système immunitaire/immunologie , Analyse de survie
7.
CNS Oncol ; 7(3): CNS22, 2018 07 01.
Article de Anglais | MEDLINE | ID: mdl-30157683

RÉSUMÉ

AIM: ERC1671 is an allogeneic/autologous therapeutic glioblastoma (GBM) vaccine - composed of whole, inactivated tumor cells mixed with tumor cell lysates derived from the patient and three GBM donors. METHODS: In this double-blinded, randomized, Phase II study bevacizumab-naive patients with recurrent GBM were randomized to receive either ERC1671 in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) (Leukine® or sargramostim) and cyclophosphamide plus bevacizumab, or placebo plus bevacizumab. Interim results: Median overall survival (OS) of patients treated with ERC1671 plus bevacizumab was 12 months. In the placebo plus bevacizumab group, median OS was 7.5 months. The maximal CD4+ T-lymphocyte count correlated with OS in the ERC1671 but not in the placebo group. CONCLUSION: The addition of ERC1671/GM-CSF/cyclophosphamide to bevacizumab resulted in a clinically meaningful survival benefit with minimal additional toxicity.


Sujet(s)
Antinéoplasiques immunologiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Bévacizumab/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Lymphocytes T CD4+/anatomopathologie , Glioblastome/traitement médicamenteux , Immunomodulation , Sujet âgé , Tumeurs du cerveau/anatomopathologie , Cyclophosphamide/usage thérapeutique , Méthode en double aveugle , Femelle , Glioblastome/anatomopathologie , Facteur de stimulation des colonies de granulocytes et de macrophages/usage thérapeutique , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Récidive tumorale locale , Résultat thérapeutique
8.
Vaccines (Basel) ; 3(1): 148-71, 2015 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-26344951

RÉSUMÉ

The existence of pathogens that escape recognition by specific vaccines, the need to improve existing vaccines and the increased availability of therapeutic (non-infectious disease) vaccines necessitate the rational development of novel vaccine concepts based on the induction of protective cell-mediated immune responses. For naive T-cell activation, several signals resulting from innate and adaptive interactions need to be integrated, and adjuvants may interfere with some or all of these signals. Adjuvants, for example, are used to promote the immunogenicity of antigens in vaccines, by inducing a pro-inflammatory environment that enables the recruitment and promotion of the infiltration of phagocytic cells, particularly antigen-presenting cells (APC), to the injection site. Adjuvants can enhance antigen presentation, induce cytokine expression, activate APC and modulate more downstream adaptive immune reactions (vaccine delivery systems, facilitating immune Signal 1). In addition, adjuvants can act as immunopotentiators (facilitating Signals 2 and 3) exhibiting immune stimulatory effects during antigen presentation by inducing the expression of co-stimulatory molecules on APC. Together, these signals determine the strength of activation of specific T-cells, thereby also influencing the quality of the downstream T helper cytokine profiles and the differentiation of antigen-specific T helper populations (Signal 3). New adjuvants should also target specific (innate) immune cells in order to facilitate proper activation of downstream adaptive immune responses and homing (Signal 4). It is desirable that these adjuvants should be able to exert such responses in the context of mucosal administered vaccines. This review focuses on the understanding of the potential working mechanisms of the most well-known classes of adjuvants to be used effectively in vaccines.

9.
Vaccine ; 33(23): 2690-6, 2015 May 28.
Article de Anglais | MEDLINE | ID: mdl-25865468

RÉSUMÉ

Glioblastoma multiforme (GBM) patients have a poor prognosis. After tumor recurrence statistics suggest an imminent death within 1-4.5 months. Supportive preclinical data, from a rat model, provided the rational for a prototype clinical vaccine preparation, named Gliovac (or ERC 1671) composed of autologous antigens, derived from the patient's surgically removed tumor tissue, which is administered together with allogeneic antigens from glioma tissue resected from other GBM patients. We now report the first results of the Gliovac treatment for treatment-resistant GBM patients. Nine (9) recurrent GBM patients, after standard of care treatment, including surgery radio- and chemotherapy temozolomide, and for US patients, also bevacizumab (Avastin™), were treated under a compassionate use/hospital exemption protocol. Gliovac was given intradermally, together with human GM-CSF (Leukine(®)), and preceded by a regimen of regulatory T cell-depleting, low-dose cyclophosphamide. Gliovac administration in patients that have failed standard of care therapies showed minimal toxicity and enhanced overall survival (OS). Six-month (26 weeks) survival for the nine Gliovac patients was 100% versus 33% in control group. At week 40, the published overall survival was 10% if recurrent, reoperated patients were not treated. In the Gliovac treated group, the survival at 40 weeks was 77%. Our data suggest that Gliovac has low toxicity and a promising efficacy. A phase II trial has recently been initiated in recurrent, bevacizumab naïve GBM patients (NCT01903330).


Sujet(s)
Glioblastome/thérapie , Immunothérapie active/méthodes , Médecine de précision/méthodes , Vaccination/méthodes , Adulte , Sujet âgé , Animaux , Femelle , Humains , Immunothérapie active/effets indésirables , Mâle , Mésothéline , Adulte d'âge moyen , Rats , Récidive , Résultat thérapeutique , États-Unis , Vaccination/effets indésirables
10.
Perm J ; 19(2): 41-6, 2015.
Article de Anglais | MEDLINE | ID: mdl-25785641

RÉSUMÉ

Glioblastoma multiforme is a highy aggressive tumor that recurs despite resection, focal beam radiation, and temozolamide chemotherapy. ERC-1671 is an experimental treatment strategy that uses the patient's own immune system to attack the tumor cells. The authors report preliminary data on the first human administration of ERC-1671 vaccination under a single-patient, compassionate-use protocol. The patient survived for ten months after the vaccine administration without any other adjuvant therapy and died of complications related to his previous chemotherapies.


Sujet(s)
Anticorps antitumoraux/immunologie , Tumeurs du cerveau/immunologie , Lobe frontal , Glioblastome/immunologie , Récidive tumorale locale/immunologie , Vaccins/immunologie , Adulte , Anticorps antitumoraux/usage thérapeutique , Bévacizumab/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Glioblastome/traitement médicamenteux , Humains , Imagerie par résonance magnétique , Mâle , Récidive tumorale locale/traitement médicamenteux , Résultat thérapeutique , Vaccins/usage thérapeutique
11.
Curr Protoc Immunol ; 106: 2.18.1-2.18.7, 2014 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-25081910

RÉSUMÉ

Vaccine adjuvants are critical components in experimental and licensed vaccines used in human and veterinary medicine. When aiming to evoke an immune response to a purified antigen, the administration of antigen alone is often insufficient, unless the antigen contains microbial structures or has a natural particulate structure. In most cases, the rationale to use an adjuvant is obvious to the experimental immunologist or the professional vaccinologist, who is familiar with the nature of the antigen, and the aim of the vaccine to elicit a specific antibody response and/or a specific type of T cell response. In this unit, we describe protocols to formulate antigens with oil-based emulsions. Such emulsions represent a major prototype adjuvant category that is frequently used in experimental preclinical vaccines, as well as veterinary and human vaccines.


Sujet(s)
Adjuvants immunologiques , Antigènes , Huiles , Vaccins , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Animaux , Antigènes/composition chimique , Antigènes/immunologie , Antigènes/pharmacologie , Émulsions , Humains , Huiles/composition chimique , Huiles/pharmacologie , Vaccins/composition chimique , Vaccins/immunologie , Vaccins/pharmacologie
12.
Vaccine ; 32(32): 4015-24, 2014 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-24837511

RÉSUMÉ

Accumulation of firm evidence that clinically apparent cancer develops only when malignant cells manage to escape immunosurveillance led to the introduction of tumor immunotherapy strategies aiming to reprogramm the cancer-dysbalanced antitumor immunity and restore its capacity to control tumor growth. There are several immunotherapeutical strategies, among which specific active immunotherapy or therapeutic cancer vaccination is one of the most promising. It targets dendritic cells (DCs) which have a unique ability of inducing naive and central memory T cell-mediated immune response in the most efficient manner. DCs can be therapeutically targeted either in vivo/in situ or by ex vivo manipulations followed by their re-injection back into the same patient. The majority of current DC targeting strategies are based on autologous or allogeneic tumor-associated antigens (TAAs) which possess various degrees of inherent tolerogenic potential. Therefore still limited efficacy of various tumor immunotherapy approaches may be attributed, among various other mechanisms, to the insufficient immunogenicity of self-protein-derived TAAs. Based on such an idea, the use of homologous xenogeneic antigens, derived from different species was suggested to overcome the natural immune tolerance to self TAAs. Xenoantigens are supposed to differ sufficiently from self antigens to a degree that renders them immunogenic, but at the same time preserves an optimal homology range with self proteins still allowing xenoantigens to induce cross-reactive T cells. Here we discuss the concept of xenogeneic vaccination, describe the cons and pros of autologous/allogeneic versus xenogeneic therapeutic cancer vaccines, present the results of various pre-clinical and several clinical studies and highlight the future perspectives of integrating xenovaccination into rapidly developing tumor immunotherapy regimens.


Sujet(s)
Antigènes hétérophiles/administration et posologie , Vaccins anticancéreux/immunologie , Tolérance immunitaire , Immunothérapie active/méthodes , Tumeurs/thérapie , Animaux , Antigènes néoplasiques/immunologie , Cellules dendritiques/immunologie , Humains , Lymphocytes T/immunologie
13.
Cytotherapy ; 16(4): 427-39, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24280238

RÉSUMÉ

Tumor growth is controlled by natural antitumor immune responses alone or by augmented immune reactivity resulting from different forms of immunotherapy, which has demonstrated clinical benefit in numerous studies, although the overall percentage of patients with durable clinical responses remains limited. This is attributed to the heterogeneity of the disease, the inclusion of late-stage patients with no other treatment options and advanced tumor-associated immunosuppression, which may be consolidated by certain types of chemotherapy. Despite variable responsiveness to distinct types of immunotherapy, therapeutic cancer vaccination has shown meaningful efficacy for a variety of cancers. A key step during cancer vaccination involves the appropriate modeling of the functional state of dendritic cells (DCs) capable of co-delivering four critical signals for proper instruction of tumor antigen-specific T cells. However, the education of DCs, either directly in situ, or ex vivo by various complex procedures, lacks standardization. Also, it is questioned whether ex vivo-prepared DC vaccines are superior to in situ-administered adjuvant-guided vaccines, although both approaches have shown success. Evaluation of these variables is further complicated by a lack of consensus in evaluating vaccination clinical study end points. We discuss the role of signals needed for the preparation of classic in situ and modern ex vivo DC vaccines capable of proper reprogramming of antitumor immune responses in patients with cancer.


Sujet(s)
Vaccins anticancéreux/usage thérapeutique , Immunité innée/effets des médicaments et des substances chimiques , Immunothérapie , Tumeurs/immunologie , Adjuvants immunologiques/usage thérapeutique , Antigènes néoplasiques/immunologie , Cellules dendritiques/immunologie , Humains , Tolérance immunitaire/immunologie , Tumeurs/anatomopathologie , Tumeurs/thérapie , Lymphocytes T/immunologie
14.
Crit Rev Immunol ; 33(6): 489-547, 2013.
Article de Anglais | MEDLINE | ID: mdl-24266347

RÉSUMÉ

Dendritic cells (DCs) are the most potent professional antigen-presenting cells, capable of initiating proper adaptive immune responses. Although tumor-infiltrating DCs are able to recognize cancer cells and uptake tumor antigens, they often have impaired functions because of the immunosuppressive tumor milieu. Therefore, DCs are targeted by therapeutic means either in vivo or ex vivo to facilitate tumor antigen presentation to T cells and induce or promote efficient antitumor immune responses in cancer patients. This immunotherapeutical approach is defined as specific active tumor immunotherapy or therapeutic cancer vaccination. In this review we briefly discuss general aspects of DC biology, followed by a thorough description of the current knowledge and optimization trends of DC vaccine production ex vivo, including various approaches for the induction of proper DC maturation and efficient loading with tumor antigens. We also discuss critical clinical aspects of DC vaccine application in cancer patients, including protocols of administration (routes and regimens), individualization of tumor immunotherapy, prediction and proper evaluation of immune and clinical responses to immunotherapy, and the critical role of combining tumor immunotherapy with other cancer treatment strategies to achieve maximal therapeutic effects.


Sujet(s)
Vaccins anticancéreux , Cellules dendritiques/immunologie , Immunothérapie/méthodes , Animaux , Présentation d'antigène , Antigènes néoplasiques/immunologie , Essais cliniques comme sujet , Cellules dendritiques/transplantation , Humains
15.
Innate Immun ; 19(1): 98-111, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-22732734

RÉSUMÉ

Dendritic cells (DCs) comprise a heterogeneous population of cells that play a key role in initiating, directing and regulating adaptive immune responses, including those critically involved in tumor immunosurveillance. As a riposte to the central role of DCs in the generation of antitumor immune responses, tumors have developed various mechanisms which impair the immunostimulatory functions of DCs or even instruct them to actively contribute to tumor growth and progression. In the first part of this review we discuss general aspects of DC biology, including their origin, subtypes, immature and mature states, and functional plasticity which ensures a delicate balance between active immune response and immune tolerance. In the second part of the review we discuss the complex interactions between DCs and the tumor microenvironment, and point out the challenges faced by DCs during the recognition of tumor Ags. We also discuss the role of DCs in tumor angiogenesis and vasculogenesis.


Sujet(s)
Cellules dendritiques/immunologie , Monitorage immunologique , Tumeurs/immunologie , Animaux , Communication cellulaire , Humains , Tolérance immunitaire , Immunité , Néovascularisation pathologique , Microenvironnement tumoral
16.
Oncoimmunology ; 1(3): 298-305, 2012 May 01.
Article de Anglais | MEDLINE | ID: mdl-22737605

RÉSUMÉ

The efficacy of immunotherapeutic TLR7/8 activation by resiquimod (R848) was evaluated in vivo, in the CNS-1 rat glioma model syngeneic to Lewis rats. The immune treatment was compared with cytotoxic cyclophosphamide chemotherapy, and as well, was compared with the combination cytotoxic and immunotherapeutic treatments. We found that parenteral treatment with the TLR7/8 agonist, resiquimod, eventually induced complete tumor regression of CNS-1 glioblastoma tumors in Lewis rats. Cyclophosphamide (CY) treatment also resulted in dramatic CNS-1 remission, while the combined treatment showed similar antitumor effects. The resiquimod efficacy appeared not to be associated with direct injury to CNS-1 growth, while CY proved to exert tumoricidal cytotoxicity to the tumor cells. Rats that were cured by treatment with the innate immune response modifier resiquimod proved to be fully immune to secondary CNS-1 tumor rechallenge. They all remained tumor-free and survived. In contrast, rats that controlled CNS-1 tumor growth as a result of CY treatment did not develop immune memory, as demonstrated by their failure to reject a secondary CNS-1 tumor challenge; they showed a concomittant outgrowth of the primary tumor upon secondary tumor exposure. Rechallenge of rats that initially contained tumor growth by combination chemo-immunotherapy also failed to reject secondary tumor challenge, indicating that the cytotoxic effect of the CY likely extended to the endogenous memory immune cells as well as to the tumor. These data demonstrate strong therapeutic antitumor efficacy for the immune response modifier resiquimod leading to immunological memory, and suggest that CY treatment, although effective as chemotherapeutic agent, may be deleterious to maintenance of long-term antitumor immune memory. These data also highlight the importance of the sequence in which a multi-modal therapy is administered.

17.
J Clin Cell Immunol ; Suppl 5: 004, 2012.
Article de Anglais | MEDLINE | ID: mdl-24955288

RÉSUMÉ

The efficacy of a various immunotherapeutic immunisation strategies for malignant glioma brain cancer was evaluated in the syngeneic CNS-1 Lewis rat glioma model. A prototype glioma cancer vaccine, which was composed of multivalent antigens derived from allogeneic and syngeneic cells and lysates, formed the prototype preparation of antigens. These antigens reflect the autologous antigens derived from the patient's surgically removed tumor tissue, as well as allogeneic antigens form glioma tumor tissue surgically removed from donor patients. This antigen mixture provides a broad spectrum of tumor associated antigens (TAA) and helps to prevent escape of tumor immune surveillance when given as a vaccine. This antigen preparation was administered in a therapeutic setting with distinct single or multiple co-stimulation-favouring immunostimulants and evaluated for inhibition of tumor growth. Our prototype vaccine was able to arrest progression of tumor growth when co-delivered in a specific regimen together with the costimulating multi-TLR agonist, Bacille Calmette Guerin (BCG) and interleukin-2, or with the Toll-Like receptor (TLR) 7/8 activator resiquimod.

18.
Expert Rev Vaccines ; 10(4): 539-50, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21506650

RÉSUMÉ

Adjuvants are essential components of most clinically used vaccines. This is because the majority of nonliving vaccines are relatively poor inducers of adaptive immunity unless effective adjuvants are co-administered. Aluminum salts (alum) have been used as adjuvants with great success for almost a century and have been particularly effective at promoting protective humoral immunity. However, alum is not optimally effective for diseases where cell-mediated immunity is required for protection. Furthermore, adjuvants including oil-in-water emulsions have shown improved efficacy for avian influenza protection suggesting that even for diseases where humoral immunity can confer protection, there is scope for developing improved adjuvants. There have been major developments in antigen discovery over the past decade, which has accelerated the vaccine development process for new indications and this demands a new generation of adjuvants that can drive and specifically direct the desired immune responses. A number of systems are under investigation that combine different types of adjuvants into specific formulations with greater activity. Additionally, targeting of vaccines to specific immune cells shows great promise. In the case of cancer and chronic infectious diseases, it may be difficult to develop effective vaccines without blocking immune regulatory pathways, which impede cell-mediated responses. However, increased understanding of immunology and particularly the innate immune system is informing vaccine adjuvant research and consequently driving the development of novel and specifically directed vaccine adjuvant strategies. In this article we address the importance of adjuvants in vaccine development, the known mode of action of specific adjuvants and recent developments in this important field.


Sujet(s)
Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , Recherche biomédicale/tendances , Contrôle des maladies transmissibles , Humains , Tumeurs/thérapie , Vaccins/immunologie
19.
PLoS One ; 6(1): e15986, 2011 Jan 31.
Article de Anglais | MEDLINE | ID: mdl-21305002

RÉSUMÉ

BACKGROUND: The innate immune system relies upon a wide range of germ-line encoded receptors including a large number of immunoglobulin superfamily (IgSF) receptors. Different Ig-like immune receptor families have been reported in mammals, birds, amphibians and fish. Most innate immune receptors of the IgSF are type I transmembrane proteins containing one or more extracellular Ig-like domains and their regulation of effector functions is mediated intracellularly by distinct stimulatory or inhibitory pathways. METHODOLOGY/PRINCIPAL FINDINGS: Carp SITR was found in a substracted cDNA repertoire from carp macrophages, enriched for genes up-regulated in response to the protozoan parasite Trypanoplasma borreli. Carp SITR is a type I protein with two extracellular Ig domains in a unique organisation of a N-proximal V/C2 (or I-) type and a C-proximal V-type Ig domain, devoid of a transmembrane domain or any intracytoplasmic signalling motif. The carp SITR C-proximal V-type Ig domain, in particular, has a close sequence similarity and conserved structural characteristics to the mammalian CD300 molecules. By generating an anti-SITR antibody we could show that SITR protein expression was restricted to cells of the myeloid lineage. Carp SITR is abundantly expressed in macrophages and is secreted upon in vitro stimulation with the protozoan parasite T. borreli. Secretion of SITR protein during in vivo T. borreli infection suggests a role for this IgSF receptor in the host response to this protozoan parasite. Overexpression of carp SITR in mouse macrophages and knock-down of SITR protein expression in carp macrophages, using morpholino antisense technology, provided evidence for the involvement of carp SITR in the parasite-induced NO production. CONCLUSION/SIGNIFICANCE: We report the structural and functional characterization of a novel soluble immune-type receptor (SITR) in a teleost fish and propose a role for carp SITR in the NO-mediated response to a protozoan parasite.


Sujet(s)
Carpes (poisson)/parasitologie , Monoxyde d'azote/immunologie , Récepteurs immunologiques/immunologie , Animaux , Carpes (poisson)/immunologie , Poissons , Immunité innée , Macrophages/parasitologie , Souris , Récepteurs immunologiques/composition chimique , Solubilité , Trypanosoma/immunologie
20.
Methods Mol Biol ; 626: 1-14, 2010.
Article de Anglais | MEDLINE | ID: mdl-20099117

RÉSUMÉ

In recent times vaccine adjuvants, or immunopotentiators, received abundant attention in the media as critical ingredients of current and future vaccines. Indeed, vaccine adjuvants are recognized to make the difference between competing vaccines based on identical antigens. Moreover, it is recognized that vaccines designed for certain indications require a matching combination of selected antigen(s) together with a critical immunopotentiator that selectively drives the required immune pathway with minimal adverse reactions. Recently, the mechanistic actions of some immunopotentiators have become clearer as a result of research focused on innate immunity receptors. These insights enable more rational adjuvant and vaccine design, which, ideally, is based on predictable immunophenotypes following vaccination.This chapter addresses immunopotentiators, classed according to their (presumed) mechanisms of action. They are categorized functionally in two major groups as facilitators of signal 1 and/or signal 2. The mode(s) of action of some well-known adjuvant prototypes is discussed in the context of this classification.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Conception de médicament , Vaccins/immunologie , Animaux , Antigènes/immunologie , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE