Статті в журналах з теми "Immunité humorale et cellulaire"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Immunité humorale et cellulaire.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-37 статей у журналах для дослідження на тему "Immunité humorale et cellulaire".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Régent, A., G. Bussone, S. V. Kaveri, and L. Mouthon. "Auto-immunité humorale et cellulaire : de la physiologie à la pathologie." La Revue de Médecine Interne 30, no. 12 (December 2009): H1—H8. http://dx.doi.org/10.1016/s0248-8663(09)73166-2.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Williams, J. C., M. G. Peacock, and R. E. Race. "Immunisation de chiens avec des vaccins contre la fièvre Q: comparaison entre des vaccins de Coxiella burnetii de phase I, phase II et du RCM de phase I." Revue d’élevage et de médecine vétérinaire des pays tropicaux 46, no. 1-2 (January 1, 1993): 87–94. http://dx.doi.org/10.19182/remvt.9404.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Des vaccins contre la fièvre Q ont été testés sur des chiens de races croisées en utilisant des cellules entières de Coxiella burnetii inactivées à la formaline dans la phase I (CEI) ou phase II (CEII) ou le résidu obtenu par extraction par chloroforme/méthanol (RCM) de cellules en phase I. Le vaccin CEI mélangé (1 : 1) à l'adjuvant incomplet de Freund (AIF) a provoqué des réponses immunitaires humorales aux antigènes des phases I et II, comme il a été mesuré par le test de microagglutination. Le vaccin RCM mélangé (1 : 1) à l'AIF a engendré des titres d'anticorps spécifiques aux antigènes de phases I et II plus élevés que le vaccin CEI. Le vaccin CEII a produit seulement des anticorps contre des antigènes de phase II. La durée d'un érythème et d'une induration après des tests dermiques avec des antigènes de Coxiella burnetii, fait penser à une immunité cellulaire. Les résultats des tests dermiques montrent que le vaccin RCM est le meilleur choix comparé aux vaccins CE, étant donné l'absence de formation tardive des granulomes par le premier. D'autres études seront nécessaires pour déterminer l'origine des réactions indésirables et pour évaluer l'efficacité des vaccins contre la coxiellose des chiens.
3

Karkouche, Raymond. "Auto-immunité à médiation cellulaire : lupus et connectivités." Annales de Pathologie 38, no. 1 (February 2018): 43–54. http://dx.doi.org/10.1016/j.annpat.2017.10.012.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Larbouret, Christel, Marie-Alix Poul, and Thierry Chardès. "Imiter la réponse immunitaire humorale polyclonale." médecine/sciences 35, no. 12 (December 2019): 1083–91. http://dx.doi.org/10.1051/medsci/2019216.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Les anticorps monoclonaux ont révolutionné le traitement de nombreuses maladies mais leur efficacité clinique reste limitée dans certains cas. Des associations d’anticorps se liant à une même cible (homo-combinaisons) ou à plusieurs cibles différentes (hétéro-combinaisons), mimant ainsi une réponse immunitaire humorale polyclonale, ont conduit à une amélioration thérapeutique dans des essais précliniques et cliniques, essentiellement en cancérologie et en infectiologie. Ces combinaisons augmentent l’efficacité des réponses biologiques et court-circuitent les mécanismes de résistances observés lors d’une monothérapie par anticorps. Le procédé de formulation et d’administration des combinaisons d’anticorps le plus fréquent est une formulation séparée, avec injection séquentielle de chaque anticorps « principe actif ». Alternativement, se développent des formulations combinées, où les anticorps produits séparément sont mélangés avant administration, ou produits simultanément par une lignée cellulaire unique ou un mélange de lignées cellulaires correspondant à une master-bank cellulaire polyclonale. La réglementation, la toxicité et la séquence d’injection des mélanges oligoclonaux restent des points à éclaircir et à optimiser pour un meilleur effet thérapeutique.
5

Wechsler, Janine. "Immunité à médiation humorale : vascularites cutanées et maladies bulleuses auto-immunes." Annales de Pathologie 38, no. 1 (February 2018): 31–42. http://dx.doi.org/10.1016/j.annpat.2017.11.003.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Frange, P., M. A. Frey, and G. Deschênes. "Immunité cellulaire et traitement immunosuppresseur du syndrome néphrotique idiopathique corticosensible." Archives de Pédiatrie 12, no. 3 (March 2005): 305–15. http://dx.doi.org/10.1016/j.arcped.2004.12.015.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Lafaix, Ch, Ch Blondeau, S. Djebbour-Levy, V. Naslet-Bonnotte, F. Faibis, and J. P. Emond. "Immunité naturelle humorale vis-à-vis des méningocoques A et C en France." Médecine et Maladies Infectieuses 25 (April 1995): 636–41. http://dx.doi.org/10.1016/s0399-077x(05)80860-1.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Kostine, Marie, Aurélien Marabelle, Thierry Schaeverbeke, and Maria Kfoury. "Les limites des inhibiteurs de points de contrôle immunitaire et la gestion de leur toxicité." médecine/sciences 35, no. 12 (December 2019): 949–56. http://dx.doi.org/10.1051/medsci/2019191.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
L’immunothérapie représente désormais un des piliers de la prise en charge du cancer, notamment avec l’arrivée des inhibiteurs de points de contrôle (checkpoint) immunitaire (ICI, immune checkpoint inhibitors). Ces anticorps thérapeutiques ciblent ces co-signaux inhibiteurs entre cellules tumorales ou cellules présentatrices d’antigènes et lymphocytes T, activant ou réactivant ainsi une immunité cellulaire T anti-tumorale. Mais la survenue d’une toxicité immunologique, qui peut concerner tous les organes, représente le facteur limitant dans le développement clinique de ces anticorps. La gestion de cette toxicité nécessite une collaboration étroite entre oncologues et spécialistes d’organe, et repose sur l’utilisation de corticoïdes et/ou d’autres immunosuppresseurs, avec l’objectif de contrôler la dysimmunité induite sans perdre l’efficacité anti-tumorale.
9

Cavaillon, Jean-Marc, Philippe Sansonetti, and Michel Goldman. "Jules Bordet, un homme de conviction." médecine/sciences 36, no. 8-9 (August 2020): 803–9. http://dx.doi.org/10.1051/medsci/2020135.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Docteur en médecine, bénéficiant d’une bourse du gouvernement belge, Jules Bordet vint se former au sein du laboratoire du père de l’immunité cellulaire, Elie Metchnikoff, à l’Institut Pasteur. Paradoxalement, il va y déchiffrer certains des mécanismes clés de l’immunité humorale, initialement découverte par l’école allemande. Il y décrit notamment les mécanismes qui aboutissent à la bactériolyse et l’hémolyse par l’action d’immunsérums. Même s’il favorisa le terme d’alexine, créé par Hans Buchner, c’est bien le système du complément (terme inventé par Paul Ehrlich) dont il est un des pères fondateurs. C’est pour ces travaux qu’il se verra attribué en octobre 1920 le prix Nobel de physiologie ou médecine millésimé 1919. Il identifia aussi le bacille de la coqueluche, qui porte son nom Bordetella pertussis.
10

Espi, M., X. Charmetant, T. Barba, L. Koppe, C. Pelletier, E. Kalbacher, E. Chalencon, D. Fouque, L. Juillard, and O. Thaunat. "Défaut de réponse humorale et cellulaire à la vaccination anti-SARS-CoV2 à ARNm chez les patients hémodialysés naïfs." Néphrologie & Thérapeutique 17, no. 5 (September 2021): 361. http://dx.doi.org/10.1016/j.nephro.2021.07.029.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
11

ELOIT, M. "Vaccins traditionnels et vaccins recombinants." INRAE Productions Animales 11, no. 1 (February 1, 1998): 5–13. http://dx.doi.org/10.20870/productions-animales.1998.11.1.3912.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Différents types de vaccins sont actuellement disponibles ou en cours de développement. Ils peuvent être divisés en deux catégories : vaccins vivants et vaccins inertes. Les vaccins vivants traditionnels incluent des souches atténuées par des moyens conventionnels, comme la croissance dans des conditions de culture inhabituelles (bactéries), ou dans des cellules ou des animaux vis-à-vis desquels les souches ne sont pas initialement adaptées (virus). Les nouvelles générations de vaccins vivants utilisant les techniques de recombinaison génétique (vaccins recombinants) peuvent être fabriquées par mutagénèse dirigée de gènes de virulence, ou par clonage de gènes de protéines immunogènes dans des vecteurs viraux ou bactériens qui possèdent les propriétés souhaitées d’innocuité et d’efficacité. Les vaccins inactivés conventionnels sont fabriqués par traitement des microorganismes par des agents physiques ou chimiques. Dans la mesure où les fractions immunogènes des microorganismes sont de mieux en mieux connues, elles peuvent être utilisées pour fabriquer des vaccins ne comprenant que ces fractions immunogènes majeures (vaccin subunitaires) par purification, ou par expressionin vitro de protéines (un autre type de vaccin recombinant) ou enfin synthèse chimique de peptides. Récemment, il a été démontré, chez différentes espèces, que l’inoculation directe dans le muscle d’un gène codant pour une protéine immunogène (immunisation génétique) permettait d’induire une réponse immune cellulaire et humorale. Cette méthode correspond à un dernier type de vaccin recombinant. L’immunité systémique et muqueuse obtenue après injection de vaccin vivant est comparée à celle obtenue après injection de vaccin inerte.
12

ROUSSEL, S., E. MERLOT, A. BOISSY, and C. DUVAUX-PONTER. "Le stress prénatal : état des connaissances et conséquences potentielles en élevage." INRAE Productions Animales 20, no. 1 (March 7, 2007): 81–86. http://dx.doi.org/10.20870/productions-animales.2007.20.1.3439.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Des stress vécus par une femelle pendant la gestation peuvent entraîner une modification du comportement de la progéniture comme une plus grande réactivité émotionnelle, des capacités d’apprentissage perturbées, une coordination motrice moins développée dans le jeune âge ainsi qu’un comportement sexuel féminisé pour les mâles. Ces modifications comportementales sont souvent accompagnées d’une modification du fonctionnement de l’axe corticotrope, avec en particulier un taux de base de glucocorticoïdes plus élevé ou des sécrétions plus importantes après un événement stressant. D’autres conséquences non négligeables sont mises en évidence comme une modification de la réponse immunitaire humorale et cellulaire de la portée, ainsi qu’une diminution du poids à la naissance. Cependant, une forte variabilité des effets est observée qui serait en partie liée à la nature de l’agent stressant, à son intensité, ainsi qu’à sa période d’application et au sexe de la progéniture, les femelles semblant plus sensibles que les mâles. Le stress prénatal serait expliqué par les hormones surrénaliennes sécrétées par la mère et traversant la barrière placentaire qui influenceraient le développement neuroendocrinien de la progéniture. La modification du comportement maternel induite par les stress répétés pourrait être un autre mécanisme expliquant les effets ob-servés chez le jeune. Même si l’essentiel des études a été réalisé chez les rongeurs et les primates, les rares expérimentations chez les animaux d’élevage montrent que les effets seraient loin d’être négligeables.
13

Navarrete, Marcelo A., Kristina Heining-Mikesch, Cristina Bertinetti-Lapatki, Marcus Duehren-von Minden, Andrea Hafkemeyer, and Hendrik Veelken. "Idiotype Vaccination of Untreated Indolent B-Cell Lymphoma: Durable Objective Remissions and Association of Superior Outcome with Cellular Immune Responses." Blood 112, no. 11 (November 16, 2008): 235. http://dx.doi.org/10.1182/blood.v112.11.235.235.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Idiotype vaccination refers to active immunization of B-cell lymphoma (B-NHL) patients with the clonal immunoglobulin (Ig) expressed by the tumor cells. After systemic cytoreductive therapy, idiotype vaccination has been shown to induce specific cellular and humoral immune responses; and humoral responses in particular are associated with prolonged remission and encouraging survival rates. Conventional idiotype vaccines are composed of the entire lymphoma-derived Ig coupled to the immunogenic carrier KLH and are administered subcutaneously with adjuvant. We have developed a idiotype production strategy based on bacterial expression of the lymphoma-derived idiotype as a recombinant Fab fragment (Bertinetti et al., EJH 2006). Intradermal administration of this antigen with lipid-based adjuvant and subcutaneous coadministration of GM-CSF had excellent immunogenicity in a phase I trial of advanced, heavily pretreated B-NHL patients (Bertinetti et al., Cancer Research 2006). In a subsequent phase II trial, 20 patients with untreated indolent B-NHL (14 follicular [FL], 3 nodal marginal zone [nMZL], 3 mantle cell [MCL]) and without immediate need for cytoreduction received at least 6 monthly idiotype vaccinations. No grade IV toxicities were seen, and the sole case of grade III toxicity, generalized erythema, did not preclude completion of the vaccination schedule. Prior to vaccination, 5/19 patients (26%) had decreased CD4+ and 6/19 patients (31%) low CD8+ T cells counts. Furthermore, 10/12 anti-HbS-negative patients (83%) failed to mount a detectable immune response to a conventional hepatitis B vaccine administered concomitantly to idiotype vaccination. Despite this functional immunodeficiency, 12/18 analyzed patients (66%) developed a cellular immune response to idiotype as detected by enumeration of IFNgamma-secreting cells by DC-ELISpot. The ELISpot protocol was validated by blinded interlaboratory testing (www.cimt.de). The frequency of idiotype-responding T cells increased from the 2nd to the 6th vaccination and could be effectively boostered by maintenance immunization in 3-monthly intervals. In vitro stimulation of PBMC from responding patients with idiotype induced specific proliferation of CD4+ T-cells and a shift towards a Th1 response in post-vaccination samples. In addition, 8/18 analyzed patients (44%) developed anti-idiotype IgG or IgM antibodies as assessed by ELISA, and the combined immune response rate was 85%. After a median follow-up of 34 months, 8 patients (40%) are progression-free, and 10 (50%) did not require cytoreductive therapy. Cellular immune responses were associated with superior PFS (p<0.05), and 5 of 6 non-responders eventually required cytoreductive therapy. Humoral immune responses were not related to PFS. Six patients (30%; only FL or nMZL) achieved an objective partial remission, including near-complete disappearance of a large submandibular mass and one subcutaneous lymphoma mass. All objective responders developed specific cellular immunity, but only 4 anti-idiotype antibodies. Five patients are in continuing remission for 12–49 months. Intradermal immunization with the chosen idiotype formulation has excellent immunogenicity despite a severely impaired immune function in untreated B-NHL patients. Furthermore, this is the first active immunotherapy trial showing objective and durable lymphoma responses in first-line therapy at a higher rate than expected for spontaneous remissions. In this setting, and in contrast to conventional idiotype vaccination schedules, cellular anti-idiotype immunity may play a crucial role for a favorable clinical outcome. Since passive humoral anti-lymphoma immunity can be easily transferred by infusions of commercially available monoclonal antibodies, synergistic benefit may be envisioned for an initial vaccination course aimed to prime anti-idiotype T-cells combined with subsequent passive immunotherapy.
14

Ulmer, Jeffrey B., Tong-Ming Fu, R. Randall Deck, Arthur Friedman, Liming Guan, Corrille DeWitt, Xu Liu, et al. "Protective CD4+ and CD8+ T Cells against Influenza Virus Induced by Vaccination with Nucleoprotein DNA." Journal of Virology 72, no. 7 (July 1, 1998): 5648–53. http://dx.doi.org/10.1128/jvi.72.7.5648-5653.1998.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
ABSTRACT DNA vaccination is an effective means of eliciting both humoral and cellular immunity, including cytotoxic T lymphocytes (CTL). Using an influenza virus model, we previously demonstrated that injection of DNA encoding influenza virus nucleoprotein (NP) induced major histocompatibility complex class I-restricted CTL and cross-strain protection from lethal virus challenge in mice (J. B. Ulmer et al., Science 259:1745–1749, 1993). In the present study, we have characterized in more detail the cellular immune responses induced by NP DNA, which included robust lymphoproliferation and Th1-type cytokine secretion (high levels of gamma interferon and interleukin-2 [IL-2], with little IL-4 or IL-10) in response to antigen-specific restimulation of splenocytes in vitro. These responses were mediated by CD4+ T cells, as shown by in vitro depletion of T-cell subsets. Taken together, these results indicate that immunization with NP DNA primes both cytolytic CD8+ T cells and cytokine-secreting CD4+ T cells. Further, we demonstrate by adoptive transfer and in vivo depletion of T-cell subsets that both of these types of T cells act as effectors in protective immunity against influenza virus challenge conferred by NP DNA.
15

Lifshitz, Lilach, Assaf Berger, Maayan Avneon, Moshe Mittelman, and Drorit Neumann. "Erythropoietin and Cyclophosphamide Combined Treatment Additively Enhances Immunoglobulin Production in Mice." Blood 112, no. 11 (November 16, 2008): 4913. http://dx.doi.org/10.1182/blood.v112.11.4913.4913.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Erythropoietin (EPO) is an important component in the treatment of cancer patients for improvement of cancer related anemia. EPO treatment for cancer related anemia is usually combined with chemotherapy. Cyclophosphamide (CP) is a known cytotoxic alkylating agent widely used in cancer chemotherapy. While at high doses it functions as an immunosuppressive agent, the anti-neoplastic activities of CP at low doses are attributed to enhancement of cellular and humoral immunity e.g. (Berd et al., Cancer Res; 1984). We have previously shown that EPO displays anti-neoplastic activities (Mittelman, 2001, 2004) and that EPO treatment is associated with enhancement of both the humoral and cellular immune responses (Prutchi-Sagiv 2006, Katz 2007). Here we focused on a murine model of DNP-KLH-injection, used to assess the humoral response in mice. Recently we demonstrated that administration of high doses of EPO (180U×3) to DNP-KLH-injected C57BL mice resulted in an increase in anti-DNP immunoglobulin G1 (IgG1) production. The present study was designed to examine the effect of combining low dose CP (12.5mg/kg ×2) used to achieve an anti-neoplastic activity, with a lower dose of recombinant human EPO (rHuEPO; 90U×3) on the humoral immune response of the DNP-KLH-injected mice, thus simulating the conditions of patient care. Hence, we compared anti-DNP Ig serum levels in DNP-KLH-injected C57BL mice that were treated with either EPO or CP alone, or the combination of CP and EPO (CP-EPO). CP treatment alone resulted in increased levels of serum anti-DNP IgG1 (O.D.(CP) = 0.38±0.06 vs O.D.(Non treated) = 0.18±0.064). In contrast, EPO treatment alone enhanced serum levels of IgG2 (O.D.(EPO) = 0.47±0.09 vs O.D.(Non treated) = 0.18±0.069). CP or EPO alone did not affect the total levels of anti-DNP total Ig (O.D.(EPO) = 0.37±0.07 vs O.D.(Non treated) = 0.28±0.04). Yet, the combined CP-EPO treatment resulted in increased levels of anti-DNP total Ig (O.D.(EPO+CP) = 0.48±0.05 vs O.D.(EPO or CP) = 0.37±0.04), maintaining the higher levels of IgG1 (O.D.(EPO+CP) = 0.38±0.06) and IgG2 (O.D.(EPO+CP) = 0.49±0.1). In summary, the combined CP-EPO treatment additively improved immunoglobulin production, compared to treatment with CP or EPO alone. We thus demonstrate that in context of chemotherapy treatment as usually administered in the clinic, EPO can enhance humoral immunity alongside its erythropoietic activities. Our findings emphasize the role of EPO as an immunomodulator, particularly when given as treatment in a combined therapeutic panel
16

Stover, C. K., G. P. Bansal, M. S. Hanson, J. E. Burlein, S. R. Palaszynski, J. F. Young, S. Koenig, D. B. Young, A. Sadziene, and A. G. Barbour. "Protective immunity elicited by recombinant bacille Calmette-Guerin (BCG) expressing outer surface protein A (OspA) lipoprotein: a candidate Lyme disease vaccine." Journal of Experimental Medicine 178, no. 1 (July 1, 1993): 197–209. http://dx.doi.org/10.1084/jem.178.1.197.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The current vaccine against tuberculosis, Mycobacterium bovis strain bacille Calmette-Guerin (BCG), offers potential advantages as a live, innately immunogenic vaccine vehicle for the expression and delivery of protective recombinant antigens (Stover, C.K., V.F. de la Cruz, T.R. Fuerst, J.E. Burlein, L.A. Benson, L.T. Bennett, G.P. Bansal, J.F. Young, M.H. Lee, G.F. Hatfull et al. 1991. Nature [Lond]. 351:456; Jacobs, W.R., Jr., S.B. Snapper, L. Lugosi and B.R. Bloom. 1990. Curr. Top. Microbiol. Immunol. 155:153; Jacobs, W.R., M. Tuckman, and B.R. Bloom. 1987. Nature [Lond.]. 327:532); but as an attenuated intracellular bacterium residing in macrophages, BCG would seem to be best suited for eliciting cellular responses and not humoral responses. Since bacterial lipoproteins are often among the most immunogenic of bacterial antigens, we tested whether BCG expression of a target antigen as a membrane-associated lipoprotein could enhance the potential for a recombinant BCG vaccine to elicit high-titered protective antibody responses to target antigens. Immunization of mice with recombinant BCG vaccines expressing the outer surface protein A (OspA) antigen of Borrelia burgdorferi as a membrane-associated lipoprotein resulted in protective antibody responses that were 100-1,000-fold higher than responses elicited by immunization with recombinant BCG expressing OspA cytoplasmically or as a secreted fusion protein. Furthermore, these improved antibody responses were observed in heterogeneous mouse strains that vary in their immune responsiveness to OspA and sensitivity to BCG growth. Thus, expression of protective antigens as chimeric membrane-associated lipoproteins on recombinant BCG may result in the generation of new candidate vaccines against Lyme borreliosis and other human or veterinary diseases where humoral immunity is the protective response.
17

Mendez-Gomez, Hector, Paul Castillo, Noah Jones, Sadeem Qdaisat, Frances Weidert, Duane Mitchell, and Elias Sayour. "COVD-02. ADAPTING RNA-NANOPARTICLE VACCINES FROM GLIOBLASTOMA TO SARS-COV-2." Neuro-Oncology 22, Supplement_2 (November 2020): ii21. http://dx.doi.org/10.1093/neuonc/noaa215.087.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract BACKGROUND Glioblastoma (GBM) can be an effective teacher in the war on COVID-19, as an operative vaccine for either must elicit near-immediate protective responses that overcomes disease heterogeneity and immune suppression. Current prophylactic strategies against COVID-19 utilize mRNA vaccines targeting small fragments of the SARS-CoV-2 genome, but these may not induce robust T cell responses or elicit immunity quickly enough. OBJECTIVE We sought to adapt an FDA-IND approved mRNA vaccine in GBM against COVID-19 for: 1) activation of near immediate immune responses, 2) targeting of full-length SARS-CoV-2 structural proteins, and 3) induction of bidirectional (B and T cell) adaptive immunity. METHODS We utilized a novel engineering design that layers mRNA into a lipid-nanoparticle (NP) shell (much like an onion); this allows greater packaging of mRNA per particle to quickly boost innate/adaptive immune responses against full-length glioblastoma antigens or SARS-CoV-2 structural proteins. RESULTS In small and large animal models, RNA-NPs safely mimic viremia activating the quiescent immune system in only a few hours for induction of protective immunity against its mRNA payload. RNA-NPs activate dendritic cells (DCs), upregulate critical innate gene signatures, and induce antigen-specific cellular and humoral immunity. We found that mice receiving SARS-CoV-2 spike RNA-NPs had more effector T cells after vaccination with significant memory recall expansion after in vitro re-stimulation with overlapping SARS-CoV-2 spike peptide mix. We also found increased release of MIP-1-alpha (i.e. CCL3) previously shown by our group (Mitchell et al. Nature 2015) to be responsible for Th1 mediated memory recall to infectious vaccine antigens in GBM patients. CONCLUSION SARS-CoV-2 RNA-NPs elicit memory recall response after vaccination. We have obtained FDA-IND approval (BB-19304, Sayour) in GBM with SARS-CoV-2 specific amendment (BB-20871) underway to support first-in-human trials of RNA-NPs targeting both GBM and COVID-19.
18

Kibkalo, D. V., S. B. Borovkov, N. I. Korenev, V. N. Borovkova та Kh A. Popova. "ВІДМІННОСТІ СКЛАДУ КРОВІ ПЕРИФЕРИЧНИХ І ЦЕНТРАЛЬНИХ ВЕН У СВИНЕЙ". Scientific Messenger of LNU of Veterinary Medicine and Biotechnology 18, № 3(70) (9 вересня 2016): 132–36. http://dx.doi.org/10.15421/nvlvet7031.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Among laboratory researches in animals the most widespread is the common blood test that can determine hidden changes in organs and tissues, monitor the effectiveness of therapeutic and preventive measures to predict outcome of disease. The question of the legality comparing the results of blood analysis from central veins with rates that are developed from capillary and peripheral vessels blood is studied in the article. Researches were conducted in seven pigs of ukrainian white breed in age 2–4 months. Blood was collected in the morning before feeding by puncture of ear vein, orbital sinus and cranial vena cava. From each animal were picked out 3 blood samples. For taking blood were applied the vacuum blood collection systems. In blood were determined the number of erythrocytes, leukocytes and hemoglobin by conventional methods (Kibkalo et al., 2016). From the results of leukocyte profile the major differences were found in the number of eosinophils whose content was significantly higher (r≤ 0.001) in the blood of ear veins. Should be noted that this index was much higher than the norm, which is possible associated with the pathological process. But their level in blood of the cranial vena cava and eye sinus was normal. Therefore, this question needs further study. Also found significantly lower (r ≤ 0.01) level of lymphocytes in the blood from the ear vein. In the vena cava were not found monocytes and basophils unlike eye sinus and ear veins, in the last their detected more. Based on the foregoing, it can be noted that in the blood of peripheral veins are registered larger number of granulocytes, which in future will be tissue macrophages. Neutrophils, monocytes, basophils and eosinophils have the ability to attach to the capillaries and small vessels walls. So they are providing the cellular immunity. In blood of central vessels are more lymphocytes that provides humoral immunity in the bloodstream. Perspectives of the next studies will be comparing the hematological results that are realized on automatic hematology analyzer from ear vein, eye sinus and cranial vena cava from the same piglets.
19

Sato, Kazuya, Junko Jimbo, Naoka Okamura, Takaaki Hosoki, Motohiro Shindo, Katsuya Ikuta, Yusuke Mizukami, Mikihiro Fujiya, Yoshihiro Torimoto, and Yutaka Kohgo. "A Crucial Cytotoxic Role of Anti-Idiotypic Antibody in Immunotherapy for B-Cell Neoplasms with Tumor Cell-Derived Heat Shock Protein 70." Blood 114, no. 22 (November 20, 2009): 1642. http://dx.doi.org/10.1182/blood.v114.22.1642.1642.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1642 Poster Board I-668 Background Tumor-derived heat shock proteins (HSPs), which bind to tumor-specific antigenic peptides, can be used for cancer immunotherapy. In this meeting, we previously reported that vaccination with mouse B-cell leukemia/lymphoma cell line, A20, -derived HSP70 (A20-HSP) against A20 cells in mice induces tumor-specific cellular immune responses (Sato et al. Blood 2001;98:1852-1857; Iuchi et al. Int J Hematol 2006;84:449-458) as well as A20-specific humoral immunity through complement dependent cell-cytotoxicities (CDCs) (Sato K et al., 2007, Blood 110:682a, Abstruct#2302). In the B-cell neoplasms, idiotype is known as one of the important tumor specific antigens which induce anti-idiotype cellular and humoral immune responses. A20-secreted monoclonal IgG (A20-Ig) and A20-idiotypic epitope peptide (A20-IP: DYWGQGTEL), which is derived from variable region of the heavy chain of the A20-Ig, are known as the A20-specific antigens (J Immunol. 2002; 168: 3983-91). Further analysis using A20-Ig and A20-IP enables to establish novel HSP70-based therapeutic strategies for B-cell neoplasms. The present study investigated whether immunization with A20-HSP induces anti-idiotypic antibodies, and also evaluated whether the antibodies showed CDCs. Methods A20 and syngeneic Balb/c mice (H-2Kd) were used in this study. HSP70 was purified from either A20 cells or healthy mouse liver tissue. A20-Ig was purified from A20 culture supernatants. After the subcutaneous immunization with A20-HSP, liver-derived HSP70 (control) to the healthy mice, the sera were harvested for the following experiments. To detect anti-A20-IP antibodies, the sera were assayed by ELISA to detect the specific IgG against A20-HSP, or A20-Ig. To confirm that immunization with A20-HSP induces anti-idiotypic antibodies, we analyzed the inhibitory effect (% inhibition) of A20-IP on the A20-HSP-mice sera reactivity against A20-HSP by preincubation of the sera with A20-IP or H-2Kd biding control peptide (mouse influenza hemagglutinin peptide: IYSTVASSL) by ELISA. To confirm that A20-HSP mice sera contains CDC type antibody directed to A20-IP, the CDC activity was determined by the trypan blue uptake of A20 cells after incubation with the complement and A20-HSP mice sera preincubated without or with A20-IP or control peptide. Results The IgG level in the A20-HSP mice against either A20-HSP or A20-Ig was significantly increased in comparison to that in control mice. % inhibition of A20-IP (45.7%) was significantly higher than that of control peptide (1.0%), indicating that almost half of IgG in the A20-HSP mice sera which reacts to A20-HSP recognizes A20-IP. The CDC activity of A20-HSP mice sera against A20 was markedly inhibited by preincubation of the sera with A20-IP but with control peptide. Conclusions: Immunization with A20-HSP70 induces anti-idiotype antibody and the antibody contributes a crucial role in eradication of A20 by CDC activity in mice. These findings enable to establish a novel and advantageous therapeutic strategy against B-cell neoplasms utilizing the anti-idiotypic antibody in HSP-based autologous tumor immunotherapy. Disclosures No relevant conflicts of interest to declare.
20

Sato, Kazuya, Junko Jimbo, Takaaki Hosoki, Motohiro Shindo, Katsuya Ikuta, Yoshihiro Torimoto, and Yutaka Kohgo. "A Novel Cytotoxic Mechanism by Leukemia-Specific Antibody in Immunotherapy with Leukemia Cell-Derived Heat Shock Protein 70." Blood 110, no. 11 (November 16, 2007): 2302. http://dx.doi.org/10.1182/blood.v110.11.2302.2302.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Introduction: Tumor-derived heat shock proteins (HSPs), which bind the tumor-specific antigenic peptides, are good application for cancer vaccine. We previously reported that immunotherapy using leukemia cell-derived HSPs against leukemia cell in mice prolonged survival by leukemia-specific cellular immune responses through CD8 + cytotoxic T-cell (Sato et al. Blood, 2001; Iuchi et al. Int J Hematol, 2006). We also indicated that CD4+ as well as CD8+ T-cell is indispensable for the survival prolongation (Sato et al. Blood, 2001), suggesting that humoral immune response by CD4+ T-cell also contributes to eradicate leukemia cells. Contributions of humoral immune responses, including tumor-specific antibodies or cytotoxic activities, in anti-tumor immunity induced by tumor-derived HSP-based immunotherapy remain unclear. We therefore investigated humoral immune responses against leukemia cells in the leukemia cell-derived HSP70-immunized mouse model. Methods: Balb/c mice and syngeneic A20 B-cell leukemia cell line were used in this study. HSP70 was purified from A20 cells or healthy mice liver tissue. After subcutaneous administration of A20-derived HSP70 (A20-HSP), liver-derived HSP70 (liver-HSP; control) to the healthy mice, the sera were harvested to perform following experiments. To detect anti-A20 antibodies, mean fluorescence intensity (MFI) of A20 cells with mice sera and FITC-conjugated anti-mouse-IgG was analyzed by flowcytometry. The sera were subjected to ELISA to detect the specific IgG against A20-HSP, or A20 secreted IgG (A20-Ig) as an A20-specific antigen. To investigate a contribution of A20-HSP70 specific CD4+ T-cell, expression of intracellular Th2-cytokine IL4 in the A20-HSP70 stimulated CD4+ T-cell in the HSP70-immunizaed mice was measured by flowcytometry. Complement dependent cytotoxicity (CDC) activities were determined by trypan blue uptake of mouse target cells (A20, YAC1: lymphoma, or T27A: myeloid leukemia) after incubation with mice sera and complement. Results: MIF of A20 with the sera from A20-HSP immunized mice (A20-HSP mice) was significantly higher than that from liver-HSP immunized mice (liver-HSP mice). IgG level against A20-HSP by ELISA was significantly increased in the A20-HSP mice compared with liver-HSP mice. The reactivities of A20-HSP mice sera against A20-HSP were completely lost by dissociation of the antigenic peptide from A20-HSP after ATP-treatment. Additionally, IgG level against A20-Ig in the A20-HSP mice was significantly higher than that in the liver-HSP mice, and this reactivity was blocked by preincubation of the sera with A20-idiotype derived peptide (A20-IP), which is the A20-specific peptide. A20-HSP70-reactive IL4-producing CD4 + T-cells in the A20-HSP mice are extremely more than those in the liver-HSP mice. The sera from A20-HSP mice showed no cytotoxic activity itself but showed significantly high CDC activity with complement against A20 but not to YAC-1 or T27A in vitro. Conclusions: Immunization with leukemia cell-derived HSP70 induces the leukemia-specific antibodies against peptides binding to leukemia cell-derived HSP70, including B-cell leukemia idiotypic peptide, via activation of the leukemia-specific CD4+ T-cell. In addition, leukemia-specific antibody-mediated CDC contributes to the eradication of leukemia cells. To utilize the leukemia-specific CDC activities induced by HSP-based immunotherapy would be a novel therapeutic strategy to eradicate leukemia cells in the patients with leukemia.
21

Thakur, Archana, Zaid Al-Kadhimi, Cassara Pray, Elyse N. Tomaszewski, Ritesh Rathore, Patricia A. Steele, Muneer H. Abidi, et al. "Transfer of Cellular and Humoral Anti-Tumor Immunity with Immune T Cells After Stem Cell Transplant (SCT) for Metastatic Breast Cancer Following “Vaccination” with Anti-CD3 x Anti-Her2/Neu Bispecific Antibody (Her2Bi) Armed Activated T Cells." Blood 118, no. 21 (November 18, 2011): 1914. http://dx.doi.org/10.1182/blood.v118.21.1914.1914.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1914 Novel therapeutic approaches are needed for women with metastatic breast cancer (BrCa). In our phase I clinical trial, infusions of anti-CD3 activated T cells (ATC) armed with anti-CD3 x anti-Her2/neu bispecific antibody (Her2Bi) induced specific cytotoxicity (SC) directed at SK-BR-3 breast cancer cells by fresh peripheral blood lymphocytes (PBL) and induced elevated serum levels of Th1 cytokines. In this study, we took advantage of armed ATC induced anti-tumor immune responses by infusing “immune” T cells collected by leukopheresis. We expanded “immune T cells” with anti-CD3 and IL-2, followed by a cryopreservation for multiple re-infusions of ATC after high dose chemotherapy (HDC) and autologous stem cell transplant (SCT). We asked whether this approach would transfer anti-tumor responses back into patients after HDC and SCT to accelerate the development of cellular and humoral anti-tumor responses. This strategy of “prime” with armed ATC and “boost” with ATC was performed in 5 evaluable patients. The “priming” portion involved 8 infusions of (2 infusions/week for 4 weeks) armed ATC given with daily low dose IL-2 (300,000 IU/m2/day) and twice weekly GM-CSF (250 μg/m2). Approximately 3 weeks after the armed ATC infusions, patients underwent a second leukopheresis for the collection and expansion of ATC. The expanded ATC from 6 patients at an effector:target ratio (E/T) of 25:1 exhibited specific cytotoxicity (SC) ranging from 3.7–25.8 (mean = 13.6%) directed at the SK-BR-3. Phenotyping of the ATC showed a mean of 50.2 % (25–74) CD4+ cells, 30.4 % (16.3-51.3) CD8+ cells, 11.6% (4.5-24.3) CD56+CD16+ cells, and 29.5% (10.4–41) CD4+CD25+ cells. A separate leukopheresis after G-CSF stimulation was performed to obtain CD34+ cells for the SCT. After HDC and PBSCT, 5 evaluable patients received multiple infusions with a mean total of 54×109 ATC (16–110 × 109) beginning day +1 after SCT. No G-CSF was given to accelerate engraftment. There were no dose-limiting side effects or delays in engraftment. One patient developed sepsis, multiple organ failure and recovered fully with supportive care and antibiotics. Phenotyping at 2 weeks after SCT showed the mean proportions of CD4+ and CD8+ cells to be 55.5 and 17.7%, respectively. Specific cytotoxicity (SC) directed at SK-BR-3 targets ranged from 4.7 to 70% at E/T of 25:1 up to 18 months post SCT but not against a negative control cell line-Daudi. Mean serum anti-SK-BR-3 antibody levels were 800 ng/ml preSCT and 1500, 1080 and 1360 ng/ml at 1, 2, and 3 months post SCT, respectively. In vitro anti-SK-BR-3 antibody synthesis was assessed using a new assay (Thakur et al, Cancer Immunol Immunother EPub, 2011) showed easily detectable levels of in vitro anti-SK-BR-3 antibody synthesis. The mean anti-SK-BR-3 antibody synthesis in the presence of CpG in pre-immunotherapy (Pre-IT), mid-IT, 1 months post-IT, pre-SCT and 1, 2, and 3 months post-SCT is summarized in the table. PBL produced anti-SK-BR-3 antibody pre-SCT and there was clear recovery of anti-SK-BR-3 antibody synthesis by PBL at 2 and 3 months after SCT. These data show infusions of immune ATC transferred cytotoxic T lymphocytes and humoral antibody activity directed at tumor antigens. These novel findings suggest that adaptive immunity was transferred into patients by ATC infusions and the stem cell product after myeloablation and SCT leading to rapid reconstitution of anti-tumor immunity.Time PointsCulture ConditionsMean±SD (n=3)Pre-ITCpG23±27Mid-ITCpG66±671M Post-ITCpG41±67Pre-SCTCpG92±101M Post-SCTCpG9±182M Post-SCTCpG45±273M Post-SCTCpG82±55 Disclosures: Lum: Transtarget Inc: Equity Ownership, Founder of Transtarget.
22

Maloney, David G., Barbara Pender, Erin McCarthy, and Daniel P. Gold. "FCgamma Receptor Polymorphisms Do Not Influence the Outcome of Treatment with Rituximab Followed by Active Immunotherapy with Mitumprotimut-T (FavId®, Id-KLH)." Blood 110, no. 11 (November 16, 2007): 3416. http://dx.doi.org/10.1182/blood.v110.11.3416.3416.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Background: Patient specific active idiotype immunotherapy with immunoglobulin idiotype is a promising new therapy for follicular NHL. Response to therapy may include both humoral and cellular anti-idiotypic immunity, but it is not clear which is most important. Prior studies have suggested that immunoglobulin FCgammaRIIIa (FCgRIIIa) polymorphisms at position 158 valine (V) or phenylalanine (F) effect the response to treatment with rituximab as well as outcomes from idiotype immunotherapy following objective response to chemotherapy. Here we present data assessing the correlation of FCgRIIIa polymorphisms and outcomes from idiotype immunotherapy following treatment with rituximab. Treatment: We determined the FCgRIIIa genotype using a SSCP method with genomic DNA isolated from 55 rituximab-naïve patients treated on a Phase II trial of mitumprotimut-T (FavId®, Id-KLH) (Koc et al, Blood, 2006; 108: #691). Four patients who progressed following rituximab and therefore did not receive mitumprotimut-T were excluded from this analysis. All 55 patients in this analysis had follicular NHL with a median age of 55 years. Thirty five patients were treatment naïve and 20 had relapsed following prior chemotherapy. Patients received rituximab (375mg/m2 i.v. weekly x 4) and those with stable or responding disease assessed at Week 11 received Id-KLH (1 mg s.q. monthly x 6) starting on Week 12 along with Leukine® (sargramostim, GM-CSF, 250 mcg, s.q.) on Days 1–4. Pts continued to receive booster injections on a reduced schedule, every other month x6 then quarterly thereafter, until disease progression. Radiological scans were performed every 3 months for the first 2 years of follow up, then every 6 months thereafter and reviewed centrally. Objective response and time to tumor progression (TTP) were assessed using modified IWG criteria (Cheson et al, J Clin Oncol1999; 17:1244). Response at 3 months, best response, TTP and progression free survival (PFS) at 1 year and 3.5 years were all assessed with respect to FCgRIIIa genotypes. Results: DNA was isolated from all 55 patients and successfully analyzed by SSCP for polymorphisms at position 158 of FCgRIIIa. Nine of 55 patients were V/V (16%), 27 were F/F (49%) and 19 were heterozygous V/F (35%). Overall, the 3 month response rate CR+PR) was 31/55 (56%) and the best overall response rate was 39/55 (71%). The 3 month response (post rituximab) was 5/9 (56%) for V/V, 9/19 (47%) for V/F and 17/27 (63%) for F/F patients. Best response was 6/9 (67%) for V/V, 12/19 (63%) for V/F and 21/27 (78%) for F/F patients. Median TTP was 19.5 months for V/V, 22.3 months for V/F and 18 months for F/F patients. The PFS at 1 year post initiation of rituximab was 57% for V/V, 61% for VF and 68% for FF patients while at the median follow-up of 3.5 years the PFS was 31% for V/V, 42% for V/F and 31% for F/F patients. Conclusions: FCgRIIIa polymorphisms were not associated with response rate or time to progression following a treatment program consisting of single agent rituximab followed by idiotype vaccination with mitumprotimut-T in rituximab-naïve patients. Results from an ongoing randomized Phase III study will assess the efficacy of this combined therapy, but these data suggest that long term PFS in patients receiving an idiotype vaccine following rituximab may rely more on a cell mediated immune response rather than a humoral response to idiotype.
23

Tsutsumi, Kazuhito, Toshiro Takafuta, Michiko Kida, Tatsuji Mino, Miki Kido, Masayuki Shimojima, Masayuki Saijo, and Takuo Ito. "Effects of Plasma Exchange (PE) Therapy in a Severe Fever with Thrombocytopenia Syndrome (SFTS) Patient with High Virus Quantities." Blood 128, no. 22 (December 2, 2016): 4900. http://dx.doi.org/10.1182/blood.v128.22.4900.4900.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Background: Severe fever with thrombocytopenia syndrome (SFTS) is an infectious disease caused by the SFTS bunyavirus, which is carried by ticks, and presents various symptoms such as fever, fatigue, digestive symptoms, and hemophagocytic lymphhistiocytosis (HLH). Since the first case was reported from China in 2011, there has been a rapid increase of newly diagnosed patients in Japan. Because the mortality rate has remained high (10-30%), there is an urgent need to establish a management strategy for SFTS. Method: We retrospectively reviewed two newly-diagnosed cases of severe SFTS in our hospital from April 2015 to July 2015. Diagnosis of SFTS was confirmed rapidly using a conventional one-step RT-PCR (cvPCR) method, and the quantity of SFTS virus in each serum was determined with a quantitative one-step RT-PCR (qPCR) method described by Yoshikawa et al. (JCM2014, 52, 3325-33). Virus titer is indicated in units of log10 copies/ml. Results: Case 1 was an 87-year-old female and Case 2 was an 81-year-old male. They showed similar severe clinical symptoms including disturbance of consciousness and a variety of abnormal laboratory findings. Both cases were quickly confirmed as SFTS using cvPCR on the next day after blood sampling. Bone marrow (BM) aspiration was performed at diagnosis, and confirmed HLH in both cases. [Case 1] Although methylprednisolone (mPSL) 250 mg was administrated to control HLH on day 3, the data indicated that HLH had become worse on day 4. To control HLH, mPSL pulse therapy (1000 mg/day) was administrated from day four to day six. However, multiple organ failure proceeded rapidly, and she expired on day eight. Serum virus quantities increased markedly from 6.99 (day 3) to 9.05 (day 6). In this case, the virus level at diagnosis was extremely high compared to previously reported cases and it was possible that the high-dose mPSL therapy suppressed anti-virus immunity. [Case 2] Although mPSL 500 mg was administrated from day two, organ damage progressed on day three. In this case, instead of increasing the dosage of mPSL, plasma exchange (PE) was performed from day four to day six, and organ damage was relieved and the patient recovered in good condition. The serum virus quantities decreased slowly from 6.62 (day 4) to 6.48 (day 7) after PE. Discussion: Yoshikawa et al. reported that virus level at diagnosis correlated to the outcomes of SFTS patients and that in most fatal cases virus quantities at diagnosis were over 5.0. In our cases, the patients had higher virus quantities at diagnosis and presented with severe symptoms. In the first case of fatality, the virus quantities remarkably increased shortly after mPSL therapy. As most SFTS patients are over the age of 50 and the mortality rate is higher in the elderly, lowered immunity in elderly patients might be the cause of SFTS progression. Moreover, immunosuppression therapy, including high dose mPSL therapy for HLH, might enhance the accelerated proliferation of SFTS virus. Although mPSL therapy is effective in some cases, immunosuppressive therapy for a SFTS patient is still controversial. In the second case, after PE therapy the virus level didn't increase and symptoms were improved. PE therapy does not suppress humoral and cellular immunity, which is different from mPSL therapy. As such, PE therapy might be effective in serious cases of SFTS with high virus quantities. In these cases, treatment plans were developed without information on virus quantities, and we obtained qPCR results afterwards. In the future, if qPCR can be performed as quickly as cvPCR, then data on the virus level at diagnosis may be the most important information for planning treatment. Disclosures No relevant conflicts of interest to declare.
24

Monath, T. P., I. Levenbook, K. Soike, Z. X. Zhang, M. Ratterree, K. Draper, A. D. T. Barrett, et al. "Chimeric Yellow Fever Virus 17D-Japanese Encephalitis Virus Vaccine: Dose-Response Effectiveness and Extended Safety Testing in Rhesus Monkeys." Journal of Virology 74, no. 4 (February 15, 2000): 1742–51. http://dx.doi.org/10.1128/jvi.74.4.1742-1751.2000.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
ABSTRACT ChimeriVax-JE is a live, attenuated recombinant virus prepared by replacing the genes encoding two structural proteins (prM and E) of yellow fever 17D virus with the corresponding genes of an attenuated strain of Japanese encephalitis virus (JE), SA14-14-2 (T. J. Chambers et al., J. Virol. 73:3095–3101, 1999). Since the prM and E proteins contain antigens conferring protective humoral and cellular immunity, the immune response to vaccination is directed principally at JE. The prM-E genome sequence of the ChimeriVax-JE in diploid fetal rhesus lung cells (FRhL, a substrate acceptable for human vaccines) was identical to that of JE SA14-14-2 vaccine and differed from sequences of virulent wild-type strains (SA14 and Nakayama) at six amino acid residues in the envelope gene (E107, E138, E176, E279, E315, and E439). ChimeriVax-JE was fully attenuated for weaned mice inoculated by the intracerebral (i.c.) route, whereas commercial yellow fever 17D vaccine (YF-Vax) caused lethal encephalitis with a 50% lethal dose of 1.67 log10 PFU. Groups of four rhesus monkeys were inoculated by the subcutaneous route with 2.0, 3.0, 4.0, and 5.0 log10PFU of ChimeriVax-JE. All 16 monkeys developed low viremias (mean peak viremia, 1.7 to 2.1 log10 PFU/ml; mean duration, 1.8 to 2.3 days). Neutralizing antibodies appeared between days 6 and 10; by day 30, neutralizing antibody responses were similar across dose groups. Neutralizing antibody titers to the homologous (vaccine) strain were higher than to the heterologous wild-type JE strains. All immunized monkeys and sham-immunized controls were challenged i.c. on day 54 with 5.2 log10 PFU of wild-type JE. None of the immunized monkeys developed viremia or illness and had mild residual brain lesions, whereas controls developed viremia, clinical encephalitis, and severe histopathologic lesions. Immunized monkeys developed significant (≥4-fold) increases in serum and cerebrospinal fluid neutralizing antibodies after i.c. challenge. In a standardized test for neurovirulence, ChimeriVax-JE and YF-Vax were compared in groups of 10 monkeys inoculated i.c. and analyzed histopathologically on day 30. Lesion scores in brains and spinal cord were significantly higher for monkeys inoculated with YF-Vax. ChimeriVax-JE meets preclinical safety and efficacy requirements for a human vaccine; it appears safer than yellow fever 17D vaccine but has a similar profile of immunogenicity and protective efficacy.
25

Hochberg, Jessica C., Jaclyn Basso, Liana Klejmont, Lauren Harrison, Allyson M. Flower, Aliza Gardenswartz, Qiuhu Shi, Perry Gerard, Humayan Islam, and Mitchell S. Cairo. "Reducing the Burden of Chemoradiotherapy with the Combination of Brentuximab Vedotin and Rituximab with Reduced Toxicity Chemotherapy in Children, Adolescents and Young Adults with Newly Diagnosed Hodgkin Lymphoma." Blood 134, Supplement_1 (November 13, 2019): 127. http://dx.doi.org/10.1182/blood-2019-128684.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background: Cure rates for newly diagnosed Hodgkin Lymphoma remain high through the combined use of chemoradiotherapy. (Hochberg/Cairo Cancer Journal 2018) However, this has resulted in significant adverse physical and psychosocial function that significantly impacts the quality of life among survivors, including cardiotoxicity, neurocognitive deficits, poor quality of life, and secondary malignancies. Pediatric cancer survivor studies have shown significant increases adverse health events following therapy throughout adulthood. Major risk factors for late effects include significant exposure to radiation, anthracyclines, cyclophosphamide, etoposide and bleomycin. (Bhakta et al Lancet 2017, Oeffinger et al NEJM 2006, Gibson et al Lancet Oncol 2018) Immunotherapy targeting the Reed Sternberg cell and tumor microenvironment, including regulatory B-cells, has potential to reduce the burden of traditional treatment. Brentuximab Vedotin, an anti-CD30 antibody-drug conjugate, and Rituximab, a chimeric anti-CD20 monoclonal antibody, have both shown efficacy in relapsed Hodgkin Lymphoma and Brentuximab has gained FDA approval in up front adult HL in combination with standard chemotherapy. (Younes et al JCO 2012, Younes et al Blood 2012, Connors et al NEJM 2018) We hypothesized that the addition of Brentuximab vedotin (Bv) and Rituximab (R) combined with risk adapted chemotherapy will be well tolerated and effective in children, adolescents and young adults with all stages of newly diagnosed Hodgkin lymphoma and will allow for the elimination of more toxic chemotherapies as well as need for radiation. Objectives: To evaluate the safety, tolerability and overall response rate of Brentuximab vedotin and Rituximab in combination with risk adapted chemotherapy in children, adolescents and young adults with newly diagnosed Hodgkin Lymphoma. Methods: Eligible patients 1-30 yrs with all stages of newly diagnosed classical Hodgkin Lymphoma. Patients were risk assigned as either Low (Stage IA, IIA, no bulk), Intermediate (Stage IA bulk/E, IB, IIA bulk, IIB or IIIA) and High Risk (Stage IB bulk/E, II bulk, IIIA bulk, IIIB, IV). Low risk patients were given 3 cycles of Brentuximab vedotin (1.2mg/kg) with Doxorubicin (25mg/m2), Vincristine (1.5mg/m2), Prednisone and Dacarbazine (375mg/m2) on Days 1 and 15. Intermediate and High Risk patients received 4 or 6 cycles of Brentuximab vedotin, Doxorubicin, Vinblastine, Dacarbazine and Rituximab (375mg/m2) on Days 1,2 and 15, 16. (Figure 1) Early response was measured by PET/CT scan. Slow responders, defined by lack of a complete metabolic response or <80% size reduction, received an additional 2 cycles of Bv-AVD-R for Intermediate Risk or Ifosfamide/Vinorelbine for High Risk. Involved Field RT was restricted to only patients with bulk disease with SER and those not in a CR at the end of therapy. Humoral and cellular immunity was measured upon completion of therapy. Results: Total enrolled = 33 patients. Median age = 15yr (range 4-23yr). Total 12 males, 21 females. Risk Assignment = 4 low, 17 intermediate, 12 high. Toxicity = 1 episode of Gr III mucositis, 1 episode of Gr III infusion reaction to Brentuximab vedotin, 2 episode Gr III peripheral neuropathy. All 33 patients achieved a complete response for a CR = 100%. Eighteen patients (58%) achieved a rapid early response. Four patients (only 12%) have required radiation therapy to date due to the presentation of bulky disease with a slow early response. Each of these patients was in CR prior to the start of radiation therapy. At a median follow-up of 18 months, the mean±SEM IgG level, CD19 and CD3 levels were 1097±63, 325±105, and 1273±290, respectively, all within normal range. Furthermore, none of the patients developed agammaglobulinemia or required hospitalization for systemic infection during or following treatment. The EFS and OS is 100% with a mean follow up time of 1320 days (= 44 months, range 3-80). (Figure 2) Conclusions: The addition of Brentuximab vedotin and Rituximab to combination risk adapted chemotherapy (without cyclophosphamide, etoposide or bleomycin) for newly diagnosed Hodgkin Lymphoma appears to be safe in children, adolescents and young adults. Our results show significant promise with a CR rate of 100%, 58% rapid early response and significant reduction in the use of toxic chemotherapy and radiation. The EFS/OS to date is 100% with a median follow up time of greater than 3.5 years. Disclosures Cairo: Osuka: Research Funding; Miltenyi: Other: MTA; Jazz Pharmaceuticals: Other: Advisory Board, Research Funding, Speakers Bureau.
26

Harrison, Simon J., Amit S. Khot, Tsin Tsin, Andy Hsu, Karen Chen, Maureen Loudovaris, Paul Neeson, et al. "Low Dose Lenalidomide and Dexamethasone Induction Followed by Autologous Transplantation In Untreated Patients with Myeloma Is Associated with High Response Rates and Preservation of CD8, but Not CD4 or NK Cellular Immunity." Blood 118, no. 21 (November 18, 2011): 1862. http://dx.doi.org/10.1182/blood.v118.21.1862.1862.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1862 Multiple Myeloma (MM) and its treatment are associated with impaired humoral and cellular immunity (CI). Lenalidomide (Len) is thought to mediate its anti-MM effect in part via stimulation of in vivo T cell, NK cell and NKT cell activity. However, as we have previously demonstrated (Hsu et al, Blood, 2011;117:1605), the immunostimulatory effects of Len are substantially abrogated by co-administration of high doses of Dexamethasone (Dex). It is unknown whether the efficacy of the Len/Dex induction regimen is maintained in newly diagnosed MM if the dose of Dex is lowered to preserve CI. In a prospective study (PMCC HREC #05/56), we examined both anti-MM responses and CI using a regimen of low dose Len and Dex induction followed by consolidation with autologous stem cell transplant (ASCT) in previously untreated patients with MM. Methods: Twenty patients were enrolled. Induction therapy was with four 28 day (d) cycles of Len 15 mg d1 − 21 and Dex 20 mg weekly followed by hematopoietic stem cell (HSC) mobilization with cyclophosphamide 2 – 4 gm/m2 and G-CSF 10mcg/kg/day, and a melphalan 140 – 200mg/m2 conditioned ASCT. Maintenance with Rev 25mg d1-21 of a 28d cycle was commenced on d21-35 post ASCT until progression. Interim assessments of response were made post-induction, post-ASCT and on 31/05/2011 using IMWG uniform response criteria. Assessments of CI were undertaken at enrolment and again at the end of 4 cycles of induction by flow cytometric analysis of peripheral blood lymphocyte subsets (CD19+, CD3+, CD4+, CD8+, CD4+/CD25+/CD127+ Treg, CD3−/ CD16+/CD56+ NK cells), T cell proliferation to allogeneic stimulators (ratio 1:1) in a 7d mixed lymphocyte reaction (MLR) and analysis of NK function by cytotoxicity against K562 targets. All data were compared to age-matched controls, and analysed for statistical significance using a student T-test. Results: Clinical responses: All patients have completed induction and ASCT. Median age=57.5 yrs, (range 44 – 70); male= 15; IgG = 10, IgA = 6 and light chain = 4. Stage at enrolment ISS1 = 9, ISS2 = 9 and ISS3 = 2. Median harvested CD34+ cells=10.8×106 /kg (range 4.9 – 40.6). The median number of CD34+ cells infused = 4.8×106 / kg (range 2.9 – 11.5) and median time to recovery of neutrophils > 0.5 ×109/L and platelets > 20 ×109/L was 12d (range 9 – 18) and 11d (range 8 – 25) respectively. The post-induction, overall response rate was 85%, very good partial response (VGPR)=20%, partial response (PR)= 65%, stable disease =5%, and 10% were refractory. Post-autograft responses improved to complete response (CR)=10%, VGPR= 40% and PR=35%. At a median follow-up of 17 months (range 5 – 29 months), the best response achieved to date is 35% CR, 25% VGPR and 30% PR (Fig 1). Immunology: CI data is available on 19 patients. Compared-to age matched controls, patients at enrolment had reduced CD3+ (p≤0.05), CD4+ (p≤0.01) (Fig 2)and Treg (P≤0.01) numbers and reduced NK cell function (p<0.05); however CD8+ T cell, NK cell and B cell numbers were normal. Following four cycles of Len/Dex therapy, CD3+, CD4+ and Treg numbers all increased, but remained below those of normal controls. Conversely, circulating B lymphocyte numbers fell substantially (P≤0.01) and NK cell function remained significantly impaired (p<0.05) following induction therapy. However, functional analysis of CD3+ (Fig 3), CD4+ and CD8+ proliferation in an MLR at enrolment and following induction therapy was identical to that of controls. Conclusion: This novel treatment regimen of low dose Len and low dose Dex followed by ASCT in untreated patients with MM is associated with high response rates and successful HSC collection. The depth of response progressively improved following ASCT. Cellular immunology in MM patients at diagnosis shows specific reductions in CD4+ and Treg cell numbers, and NK cell function that is not rescued by Len therapy when co-administered with Dex, even at low doses, which induced a further reduction in CD19+ cells. However despite these deficiencies, preserved proliferative capacity of both CD4+ and CD8+ T-cells is seen at diagnosis and following this induction therapy, suggesting that with the low dose Len-Dex regimen, the immune environment is conducive to novel strategies that are aimed at inducing adaptive anti-MM immune responses. Disclosures: Harrison: Celgene: Honoraria, Research Funding. Off Label Use: Low dose lenalidomide in newly diagnosed myeloma. Neeson:Celgene: Research Funding. Prince:Celgene: Honoraria, Research Funding.
27

Zappasodi, Roberta, Alessandra Cavanè, Monica Tortoreto, Cristina Tringali, Giusi Ruggiero, Lorenzo Castagnoli, Bruno Venerando, et al. "HSP105 Inhibition Counteracts Key Oncogenic Pathways and Hampers the Growth of Human Aggressive B-Cell Non-Hodgkin Lymphoma." Blood 120, no. 21 (November 16, 2012): 1562. http://dx.doi.org/10.1182/blood.v120.21.1562.1562.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1562 Our previous findings have made it clear that the significant clinical efficacy attained by dendritic cell-based vaccination in relapsed B-cell non-Hodgkin lymphoma (B-NHL) patients is firmly associated with multifaceted immunologic responses, including the development of anti-heat shock protein (HSP)105 humoral immunity (Di Nicola et al., Blood 2009 113:18–27; Zappasodi et al., Cancer Res. 2010 70:9062–9072; Zappasodi et al., Blood 2011 118:4421–4430). Human HSP105 is a high-molecular-weight chaperone constitutively expressed at low levels within the cytoplasm, and can also be induced in the nucleus by various forms of stress. It is overexpressed in several solid tumors, including melanoma, breast, thyroid and gastroenteric cancers. We have recently shown that this is also true for B-NHLs, in which HSP105 levels increase in function of their aggressiveness and proliferation index (Zappasodi et al., Blood 2011 118:4421–4430). Accordingly, in normal lymph nodes HSP105 expression is confined to the hyper-proliferating germinal center (GC) B cells, suggesting its involvement in the potentially oncogenic GC reactions. We have now set out to clarify the functional role of HSP105 in B-NHLs by stably silencing its expression in the Namalwa aggressive lymphoma cell line. Namalwa cells were infected by using a lentiviral vector carrying a HSP105-targeting pre-microRNA sequence and the Emerald Green Fluorescent Protein (EmGFP) gene, both under the human cytomegolovirus immediate early promoter, as well as the blasticidin resistance gene. Control cells were mock-infected with the empty vector. Infected cells were initially selected in the presence of blasticidin, and then single GFP+ cells were sorted on a flow cytometry device. In this way, we achieved 100% GFP+ subclones that displayed a specific constitutive down-regulation of HSP105, as there was no significant decrease in the expression of its cognate molecular homolog HSP70, or the other major cellular chaperone HSP90. Comparison of the in vitro proliferation rate of two silenced clones with that of the mock culture showed that the cell doubling time of both clones significantly increased and their in vitro growth was accordingly delayed (P= 0.01 and P= 0.04). Western blot analysis in 6 different silenced clones of the oncoproteins most frequently involved in B-NHLs revealed that BCL-6 and c-Myc were down-regulated in function of HSP105 knockdown levels, whereas in mock cells no modifications were detected with respect to their wild-type counterparts. Further strengthening the association between HSP105, BCL-6 and c-Myc expression, immunohistochemistry analysis of 50 primary human aggressive B-NHLs revealed that HSP105 expression, measured both as intensity and percentage of positive cells, was significantly higher in c-Myc- or BCL-6-dependent Burkitt (P= 0.0264) and diffuse large B-cell lymphomas (P= 0.0068) respectively than in other aggressive istotypes that do not overexpress these oncoproteins. These findings support the potential pro-tumorigenic cooperation of HSP105 with BCL-6 and c-Myc transcription factors. To find out whether counteracting HSP105 functions hampers in vivo lymphoma growth, we evaluated the tumor-forming capability of HSP105-silenced (siHSP105) or mock Namalwa cells subcutaneously injected into severe combined immunodeficient mice at serial 10-fold dilutions from 106 to 104 cells/injection (Figure 1). We found that HSP105 knockdown slightly delayed in vivo Namalwa tumor formation when 106 and 105 cells were injected. Noteworthy, no lesions appeared over 70-day observation in mice inoculated with 104 siHSP105 cells, whereas palpable tumors were present in 67% of the animals 24 days after injection of the mock cells (Figure 1). Overall, these results indicate that HSP105 may be a per se nononcogenic molecule that contributes to lymphomagenesis by facilitating the tumorigenic functions of key oncoproteins. They equally provide the rationale for developing HSP105 inhibitors as a novel strategy for improving the treatment of aggressive B-NHLs. Figure 1. In vivo tumor-forming capability of siHSP105 or mock Namalwa cells Figure 1. In vivo tumor-forming capability of siHSP105 or mock Namalwa cells Disclosures: Gianni: Hoffmann-La Roche: Consultancy, Honoraria.
28

Graver, J. C., W. F. Jiemy, D. Altulea, A. Boots, P. Heeringa, W. Abdulahad, E. Brouwer, and M. Sandovici. "OP0062 CYTOKINE PRODUCING B CELLS SKEW MACROPHAGES TOWARDS A PRO-INFLAMMATORY PHENOTYPE IN GIANT CELL ARTERITIS." Annals of the Rheumatic Diseases 80, Suppl 1 (May 19, 2021): 33.1–34. http://dx.doi.org/10.1136/annrheumdis-2021-eular.1984.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background:Giant cell arteritis (GCA) is the most frequent form of systemic vasculitis affecting the temporal artery (TA) and the aorta. Macrophages and T cells are well recognized players in the pathogenesis of GCA while B cells are often not taken into account. Recently, changes in the circulating B cell compartment were documented in GCA and B cells were found to organize into tertiary lymphoid organs at the site of vascular inflammation (TA and aorta).1,2 The exact role of B cells in GCA is still unknown as no disease-specific antibodies have been defined. However, beside their role in humoral immunity, B cells can also produce various cytokines. In GCA, peripheral B cells of treated GCA patients, showed an enhanced capacity to produce pro-inflammatory cytokine Interleukin (IL)63 which is nowadays an important target of treatment in GCA.4 We hypothesize that B cells help shape the inflammatory response in GCA by producing effector and regulatory cytokines.Objectives:We aimed to assess the cytokine profile of circulating and lesional B cells in GCA and studied the effects on macrophage skewing.Methods:To assess B cells with the capacity to produce cytokines, cryopreserved peripheral blood mononuclear cells of 11 untreated GCA patients and 15 age- and sex-matched healthy controls (HC) were cultured for 3 days in the presence of CpG-ODN 2006. During the last 5 hours phorbol myristate acetate and Calcium Ionophore were added. Thereafter, intracellular effector (IL6, TNFa, IFNy, LTb) and regulatory B cell-related cytokines (IL10) were measured with flow cytometry. To assess potential skewing of macrophages by B cell products, THP-1 cells were differentiated into macrophages and stimulated for 24 hours with supernatant from stimulated B cells (n=6). Expression of IL23, IL6, IL1b, TNFa, MMP9 and YKL40 was assessed on mRNA level with qPCR. To assess local cytokine production, TA (n=11) and aorta tissue samples (n=10) of histologically-proven GCA patients were stained to detect CD20, IL6, TNFa, IFNy, LTb, and IL10 expression. For comparison, 14 aorta tissues samples of patients with an atherosclerosis-related aortic aneurysm were included.Results:In vitro stimulated B cells from untreated GCA patients showed an enhanced percentage of IL6+ B cells (median (IQR); 44 (41-52)) and of IL6+TNFa+ B cells (12 (8-24)) compared to stimulated B cells from HC (IL6+: 28 (23-39), IL6+TNFa+: 6 (4-24)). In addition, soluble factors, secreted by GCA derived and stimulated B cells, skewed macrophages towards a pro-inflammatory phenotype with enhanced expression of IL23, IL6, IL1b, and TNFa. Furthermore, these macrophages also showed higher expression of the tissue remodelling factor MMP9 and the pro-angiogenic factor YKL40. At the site of vascular inflammation, B cells were detected in the regions with clear TNFa, IL6, IFNY, LTb and IL10 expression in both the TA and aorta of GCA patients.Conclusion:This study demonstrates that circulating B cells of patients with GCA have the capacity to express pro-inflammatory cytokines (IL6 and TNFa) which can influence other cellular players in GCA. Specifically, B cell secreted soluble factors were able to skew macrophages towards a pro-inflammatory phenotype. In addition, this study provides evidence for an active role of B cells in shaping the cytokine milieu at the site of inflammation thereby revealing the B cell as a new target of intervention in GCA.References:[1]Ciccia, F. et al. Ectopic expression of CXCL13, BAFF, April and LT-β is associated with artery tertiary lymphoid organs in giant cell arteritis. Ann. Rheum. Dis.76, 235–243 (2017)[2]Graver, JC. et al. Massive B cell Infiltration and Organization Into Artery Tertiary Lymphoid Organs in the Aorta of Large Vessel Giant Cell Arteritis. Front. Immunol.10, 83 (2019)[3]Van Der Geest, KSM. et al. Disturbed B cell homeostasis in newly diagnosed giant cell arteritis and polymyalgia rheumatica. Arthritis Rheumatol.66, 1927–1938 (2014)[4]Stone, JH. et al. Trial of tocilizumab in giant-cell arteritis. N. Engl. J. Med.377, 317–328 (2017)Disclosure of Interests:Jacoba Carolien Graver: None declared, William Febry Jiemy: None declared, Dania Altulea: None declared, Annemieke Boots Consultant of: Grunenthal, Peter Heeringa: None declared, Wayel Abdulahad: None declared, Elisabeth Brouwer Speakers bureau: Roche, fees paid to UMCG, Maria Sandovici: None declared
29

Stadtmauer, Edward A., Dan T. Vogl, Nicole A. Aqui, Aaron P. Rapoport, Kenyetta Macdonald, Heather Murphy, Rita Bhagat, et al. "Influenza (flu) Vaccine Administration to Patients with Multiple Myeloma (MM) Prior to Autologous T-Cell Harvest Leads to Better Reconstitution of Flu Immunity After High Dose Melphalan and Autologous Stem Cell Transplant (ASCT) and Reinfusion of Primed Ex-Vivo Co-Stimulated Autologous T-Cells and Post-ASCT Second Flu Vaccination Than Post-ASCT Flu Vaccination Alone." Blood 114, no. 22 (November 20, 2009): 797. http://dx.doi.org/10.1182/blood.v114.22.797.797.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 797 Introduction: Epidemics of influenza A virus strains have been associated with a mortality rate of 0.1% and hospitalization of approximately 200,000 people per year in the United States. The recent pandemic of swine-origin H1N1 has further emphasized the need to protect the population. Patients with multiple myeloma (MM) are particularly vulnerable to flu infection from altered humoral and cellular immunity from the disease as well as from the immunosuppressive chemotherapies, corticosteroids and radiation frequently used for treatment. In particular, high dose melphalan and ASCT, a common and efficacious treatment as part of first-line or salvage therapy for MM, is associated with post-ASCT immune suppression by virtue of low T-cell numbers, poor T-cell function and immune paresis. Despite anti-infection vaccination after ASCT, pneumococcal pneumonia and influenza are common. In an attempt to improve this outcome we previously showed that adoptive transfer of in-vivo PCV vaccine-primed and ex-vivo (anti-CD3/anti-CD28) co-stimulated autologous T cells (aT-cells) early (within 14 days) post-ASCT increased CD4 and CD8 T cell counts and induced pneumococcal conjugate vaccine-directed T and B-cell responses [Rapoport et al, Nature Medicine, 2005]. Patients who did not receive pre-collection vaccination did not show this response despite infusion of co-stimulated T-cells and post-ASCT vaccination. In the current clinical trial we investigated whether pre-harvest flu vaccination would result in a similar improvement in immunity. Methods: We performed a randomized open-label single center study of two influenza vaccine schedules for patients with high-risk MM responding after conventional induction therapy. Of 21 patients enrolled, 11 were randomly assigned to Group X and received pre- and d+14 post-transplant flu vaccine with the commercially available influenza vaccine (Fluzone) produced by Aventis Pasteur. The 10 patients in Group Y received only post-transplant vaccination. All patients received T-cell harvest, high dose melphalan and ASCT and aT-cell infusion. Immunological assessment (T-cell and B-cell subsets, T-cell repertoire, T-ELISPOT, CFSE-CBC, antibody HAI, B-ELISPOT) was conducted d+60, 100 and 180. The primary study endpoint is the immune response to influenza as measured by the serotype-specific influenza antibody response on day 100 using a standard hemagglutination inhibition assay (HAI) optimized for the vaccine administered each year. Results: The median age was 55 (range 37-68), 13 M, 8 F, 18 Caucasian, 2 African American, 1 Hispanic, IgG 57%, IgA 33%, light chain 10%. With a median follow-up of 1 year, 86% (18/21) patients are alive with 48% (10/21) alive in remission. No difference in these parameters was seen between both Groups. HAI titers, however were higher at all three time points (d+60, 100, 180) for Group X when compared to Group Y both for H3N2 (p= 0.04) and for H1N1 (p=0.07). HAI titers for Group Y remained near baseline throughout all time points while Group X peaked day 60 and remained elevated day 180. Analysis of other immune assessment is ongoing and will be presented. Conclusion: Preliminary analysis of this randomized trial of flu vaccine schedule for MM patients undergoing ASCT suggests superior flu immune reconstitution when vaccine is administered to prime autologous T-cells prior ASCT and aT-cell infusion. This result adds to our observation that an approach of vaccine priming and co-stimulated autologous T-cell infusion after high dose melphalan and ASCT for MM augments an otherwise poor response in this patient population to pneumococcal and influenza vaccination. Investigation of this approach as a platform for anti-myeloma immune therapy is ongoing. Disclosures: June: University of Pennsylvania: Patents & Royalties.
30

Dhedin, Nathalie, Jacques-Olivier Bay, Patricia Ribaud, Mathieu Coudert, Marie T. Rubio, Philippe Agape, Anne Thiebaut, et al. "Response to Adjuvanted Monovalent Influenza A (H1N1)v Vaccine In Allogeneic Hematopoietic Stem Cell Transplant Recipients." Blood 116, no. 21 (November 19, 2010): 1269. http://dx.doi.org/10.1182/blood.v116.21.1269.1269.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1269 Background: Influenza is a potentially serious infection after hematopoietic stem cell transplantation (HSCT). Prolonged immunosuppression leads to impaired immunity to infectious agents that contributes to the poor outcome after HSCT. Vaccination is the main prophylactic approach in individuals at an increased risk for severe influenza disease, but it is less effective in immunocompromised patients. Nevertheless, annual vaccination against influenza is recommended for HSCT recipients, starting at 6 months after transplant. In 2009, due to the emergence of a pandemic influenza A (H1N1)v virus, the development of safe and effective vaccines was a public health priority. Some oil-in-water-emulsion adjuvants were used in some 2009 influenza A (H1N1) vaccines to increase their immunogenicity. In France, the use of 2 doses of such vaccines was recommended for HSCT recipients. Methods: This study, conducted by the Société de Greffe de Moelle et de Thérapie Cellulaire, has evaluated the safety and the efficacy of an adjuvanted monovalent influenza A (H1N1)v vaccine in allogeneic HSCT recipients. Patients between the age of 18 and 65 years who were vaccinated from 3 months to 5 years post-transplant were included in the study. Patients in relapse of their hematological disease or receiving immunoglobulins were excluded. Patients were separated into two groups: patients with graft-versus-host-disease (GVHD) treated by immunosuppressive drugs (G1) and those without GVHD or immunosuppressive therapy (G2). Antibody responses were measured by means of a hemagglutination-inhibition assay on days 0, 21 and 42 after injection of the first dose of vaccine. Results: Seventy nine patients were included and 70 who received the 2 doses of adjuvanted vaccine (at day 0 and 21) were analyzed: 41 in G1, 29 in G2. Median patient age was 53 yr (range 21–65). Median interval between transplant and vaccination was11 months in G1 and 21months in G2. Fifty five % of patients received stem cells from peripheral blood and 44% received a myeloablative conditioning regimen. No severe post-vaccination side effect was observed, except in 2 patients who presented with an aggravation of their GVHD. No case of influenza A (H1N1)v infection was observed. At day 21 after the first dose, antibody titers, expressed as geometric means (GMT), were 20 and 69 in G1 and G2, respectively, whereas, they were 43 and 223, respectively, at day 42 after the second dose. By day 21, antibody titers of 1:40 or more were observed in 34% of patients in G1 and 73% in G2. By day 42, after the second dose of vaccine, antibody titers of 1:40 or more were observed in 47 % of patients in G1 and 86% in G2. The reverse cumulative distribution curves of antibody titers in serum at D0, D21 and D42 are shown in Figure 1. Conclusions: These data show that adjuvanted vaccine is safe in recipients of allogeneic HSCT. The humoral response was improved by the second dose of vaccine. The use of 2 doses of adjuvanted vaccine allows a seroprotection in almost all recipients without GVHD and immunosuppressive therapy and in half of the patients with GVHD and treated by immunosuppressive drugs. These data also suggest that addition of an adjuvant to improve the efficacy of a vaccine offers an advantage in recipients of HSCT. Disclosures: No relevant conflicts of interest to declare.
31

Melo-Cardenas, Johanna, Mauricio Urquiza, Robier Aguillon-Prada, Thomas J. Kipps, and Januario E. Castro. "Ad-ISF35 Intratumoral Administration Induces a Bystander Effect and Immune-Mediated Tumor Rejection with a Safe Vector Biodistribution and Toxicology Profile In a NHL Mouse Model." Blood 116, no. 21 (November 19, 2010): 1470. http://dx.doi.org/10.1182/blood.v116.21.1470.1470.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 1470 Ad-ISF35 is a replication-defective adenovirus (Ad) vector encoding a membrane-stable chimeric CD154 (ISF35) that can induce expression of costimulatory and death receptor molecules in chronic lymphocytic leukemia (CLL) cells in vitro and in vivo. Ad-ISF35 has shown to be safe and to have potential activity in two clinical studies in CLL patients (Wierda et al, Leukemia, 2010 in press; Castro et al. Blood 2008 112: Abstract 2100). However, the activity, immunogenicity, safety and biodistribution of Ad-ISF35 intratumoral administration have not been evaluated in a solid tumor model. To study these questions, we use BALB/c mice bearing subcutaneous A20 Non-Hodgkin Lymphoma (NHL) tumors as an in vivo cancer vaccine model. A20 tumor bearing mice were injected intratumoraly with single or repeat dose administration of Ad-ISF35 [3×109 or 3×1010 viral particles (vp)], Ad-Empty or control vehicle on days 0, 7 and 14. Treated animals were sacrificed on days 2, 5, 19 and 39. Clinical observations, comprehensive necropsy, hematology, serum chemistry, histopathology, cytokine and protein expression as well as Ad-ISF35 vector biodistribution and ISF35 transgene expression were analyzed. Ad-ISF35 injection in A20 tumor-bearing mice was well tolerated with mild toxicities that included mild and transient elevation in RBC, hemoglobin and hematocrit treated males. In both sexes we observed a transient increase in neutrophils on day 2 after Ad-ISF35 injection, and there were no abnormalities in serum chemistry suggestive of major organ dysfunction. Histopathology assessment of ten different organs did not show differences between the controls and Ad-ISF35 injected mice. Ad-ISF35 biodistribution showed that the viral vector was primarily found in the injected tumors and underwent a rapid clearance with no evidence of accumulation or persistence in the injected tumor or peripheral organs. Half of the Ad-ISF35 positive samples showed evidence of ISF35 transgene expression measured by real time q-PCR. Immunohistochemistry analysis of A20 tumors 48 hours after Ad-ISF35 intratumoral injection showed a massive infiltration of monocytes, neutrophils and T-cells as well as evidence of tumor cell apoptosis that correlated with tumor shrinkage followed by complete regression in 80% of Ad-ISF35 injected mice but not in the control animals. Following Ad-ISF35 injection, we observed CD154 upregulation in 0.25% of the tumor cells while there was a 100-fold increase in the number of cells expressing CD40 suggesting a potent bystander effect in non-transduced tumor cells. Analysis of the cytokine expression profile in tumor cells from mice treated with Ad-ISF35 intratumoral injection showed increased expression of IL-6, IFN-γ and MCP-1 as early as 48 hours after injection; this was not observed in the control mice. Mice that were cured from A20 tumors after intratumoral Ad-ISF35 injection developed antibodies and cytotoxic T cells that reacted specifically with A20 cells and prevented the development of A20 tumors after rechallenge. In summary, we show here that Ad-ISF35 intratumoral injection in a solid tumor NHL mouse model can function as an in vivo cancer vaccine breaking tumor tolerance and inducing complete tumor regression and immunity to tumor rechallenge. Ad-ISF35 intratumoral administration was well tolerated and showed a favorable biodistribution viral vector profile with rapid clearance and no evidence of vector persistence. Ad-ISF35 intratumoral injection induced expression of immunomodulatory and chemotactic cytokines as well as humoral and cellular anti-tumoral responses. Moreover, the effect of intratumoral Ad-ISF35 administration appears to be associated with a potent bystander effect in non-transduced cells. These data provide the rationale for development of Ad-ISF35 intratumoral administration as an in vivo cancer vaccine strategy in patients with NHL and other solid tumors. Disclosures: Kipps: Memgen LLC: Membership on an entity's Board of Directors or advisory committees.
32

Smith, Christof, Sarah Entwistle, Caryn Willis, Steven Vensko, Wolfgang Beck, Jason Garness, Maria Sambade, et al. "478 Translation of a therapeutic neoantigen vaccine workflow to SARS-CoV-2 vaccine development." Journal for ImmunoTherapy of Cancer 8, Suppl 3 (November 2020): A510—A512. http://dx.doi.org/10.1136/jitc-2020-sitc2020.0478.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
BackgroundThere is an urgent need for a vaccine with efficacy against SARS-CoV-2. We hypothesize that peptide vaccines containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation would drive both humoral and cellular immunity with high specificity, potentially avoiding undesired effects such as antibody-dependent enhancement (ADE) (figure 1). Leveraging methods initially developed for prediction of tumor-specific antigen targets, we combine computational prediction of T cell epitopes, recently published B cell epitope mapping studies, and epitope accessibility to select candidate peptide vaccines for SARS-CoV-2 (figure 2).MethodsSARS-CoV-2 HLA-I and HLA-II ligands were predicted using multiple MHC binding prediction software. T cell vaccine candidates were further refined by predicted immunogenicity, viral source protein abundance, sequence conservation, coverage of high frequency HLA alleles, and co-localization of CD4+/CD8+ T cell epitopes. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, filtering to select regions with surface accessibility, high sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. Using murine compatible T/B cell epitopes, vaccine studies were performed with downstream ELISA/ELISpot to monitor immunogenicity.ResultsWe observed distribution of HLA-I (n = 2486) and -II (n = 3138) ligands evenly across the SARS-CoV-2 proteome, with significant overlap between predicted human and murine ligands (figure 3). Applying a multivariable immunogenicity model trained from IEDB viral tetramer data (AUC 0.7 and 0.9 for HLA-I and -II, respectively), alongside filters for entropy and protein expression resulted in 292 CD8+ and 616 CD4+ epitopes (figure 4). From an initial pool of 58 B cell epitope candidates, three epitope regions were identified (figure 5). Combining B cell and T cell analyses, alongside manufacturability heuristic, we propose a set of SARS-CoV-2 vaccine peptides for use in subsequent murine studies and clinical trials (figure 6). Preliminary murine studies demonstrate evidence of T and B cell activation (figure 7).Abstract 478 Figure 1Summary of combination CD4+/CD8+ T cell and B cell SARS-CoV-2 peptide vaccine. Humoral immunity (blue dashed box) is targeted through B cell and HLA-II epitopes, aimed at viral neutralization while avoiding non-neutralizing and ADE promoting targets. Cellular immunity (red dashed box) is targeted through HLA-I and HLA-II epitopes, aimed to clear virally infected cellsAbstract 478 Figure 2Summary of B cell and CD4+/CD8+ epitope prediction workflows. Pathways are colored by B cell (blue), human T cell (black), and murine T cell (red) epitope prediction workflows. Color bars represent proportions of epitopes derived from internal proteins (ORF), nucleocapsid phosphoprotein, and surface-exposed proteins (spike, membrane, envelope)Abstract 478 Figure 3Landscape of SARS-CoV-2 MHC ligands. (A&B) Selection criteria for (A) HLA-I and (B) HLA-II SARS-CoV-2 HLA ligand candidates. Scatterplot (bottom) shows predicted (x-axis) versus IEDB (y-axis) binding affinity, with horizontal line representing 500 nM IEDB binding affinity and vertical line representing corresponding predicted binding affinity for 90% specificity in binding prediction. Histogram (top) shows all predicted SARS-CoV-2 HLA ligand candidates. (C) Landscape of predicted HLA ligands, showing nested HLA ligands comprising HLA-I and -II ligands with complete overlap (top), and LOESS fitted curve (span = 0.1) for HLA-I/II ligands by location along the SARS-CoV2 proteome (bottom). Red track represents SARS epitopes identified in literature review with sequence identity in SARS-CoV-2. Predicted HLA ligands with conserved sequences to this literature set are represented in the lollipop plot with a red stick. (D) Summary of total number of predicted HLA-I/II ligands and nested HLA ligands. (E) Summary of nested HLA ligand coverage by protein, with raw counts (left) or counts normalized by protein length (right). (F) Summary of murine/human MHC ligand overlap. (G) Distribution of population frequencies among predicted HLA-I, -II, and nested HLA ligandsAbstract 478 Figure 4Prediction of SARS-CoV-2 T cell epitopes. (Top) Summary of predicted (left) and IEDB-defined (right) SARS-CoV-2 HLA ligands, showing proportions of each derivative protein. (Middle) Funnel plot representing counts of HLA-I (red text), HLA-II (blue text), and nested HLA (violet text) ligands along with proportions of HLA-I (top bar) and HLA-II (bottom bar) alleles at each filtering step. (Bottom) Summary of CD8+ (red, top), CD4+ (blue, bottom), and nested T cell epitopes (middle) after filtering criteria in S, M, and N proteins. Y-axis and size represent the population frequency of each CD8+ and CD4+ epitopes by circles. Middle track of diamonds represents overlaps between CD8+ and CD4+ epitopes, showing the overlap with greatest population frequency (size) for each region of overlap. Color of diamonds represents the proportion of overlap between CD4+ and CD8+ epitope sequences.Abstract 478 Figure 5Selection of SARS-CoV-2 B cell epitope regions. (A) SARS-CoV-2 linear B cell epitopes curated from epitope mapping studies. X-axis represents amino acid position along the SARS-CoV-2 spike protein, with labeled start sites. (B) Schematic for filtering criteria of B cell epitope candidates. (C) Spike protein amino acid sequence, with overlay of selection features prior to filtering. Polymorphic residues are red, glycosites are blue, accessible regions highlighted in yellow. The receptor binding domain (RBD), fusion peptide (FP), and HR1/HR2 regions are outlined. (D) Spike protein functional regions (RBD, FP, HR1/2) amino acid sequences, with residues colored by how many times they occur in identified epitopes. Selected accessible sub-sequences of known antibody epitopes highlighted in purple outline. (E) S protein trimer crystal structure with glycosylation, with final linear epitope regions highlighted by colorAbstract 478 Figure 6T cell and B cell vaccine candidates. (A) 27mer vaccine peptide sets selecting for best CD4+, CD8+, CD4+/CD8+, and B cell epitopes with HLA-I, HLA-II, and total population coverage. (B) Unified list of all selected 27mer vaccine peptides. Vaccine peptides containing predicted ligands for murine MHC alleles (H2-b and H2-d haplotypes) are indicated in their respective columnsAbstract 478 Figure 7Immunogenicity of murine-compatible peptide vaccines. (A) ELISA result: peptides derived from three B cell vaccine candidate regions were coated on peptide capture plates, either in combination by overlapping core epitopes (1+2 and 3+4) or alone (5). (B) ELISpot results: splenocytes from animals vaccinated against predicted B cell epitopes (1–5) or measles peptide control (M; adapted from Obeid et al. 1995). Each point represents the average of technical triplicates, background subtracted against no-peptide control. (A&B) Colors represent adjuvant used for vaccination. P-values shown above each graph represent pair-wise Mann-Whitney u-testConclusionsA peptide vaccine targeting B cells, CD4+ T cells, and CD8+ T cells in parallel may prove an important part of a multifaceted response to the COVID-19 pandemic. Adapting methods for predicting tumor-specific antigens, we presented a set of peptide candidates with high overlap for T and B cell epitopes and broad haplotype population coverage, with validation of immunogenicity in murine vaccine studies.AcknowledgementsThe authors appreciate funding support from University of North Carolina University Cancer Research Fund (AR and BGV), the Susan G. Komen Foundation (BGV), the V Foundation for Cancer Research (BGV), and the National Institutes of Health (CCS, 1F30CA225136). We would like to thank members of the #DownWithTheCrown Slack channel for helpful discussion and feedback.
33

Pavlica, Ljiljana, Nada Pejnovic, and Nada Draskovic. "Cell-mediated immune response of synovial fluid lymphocytes to ureaplasma antigen in Reiter's syndrome." Srpski arhiv za celokupno lekarstvo 131, no. 7-8 (2003): 285–89. http://dx.doi.org/10.2298/sarh0308285p.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
INTRODUCTION Reiter's syndrome (RS) is an seronegative arthritis that occurs after urogenital or enteric infection which in addition with occular and/or mucocutaneous manifestations presents complete form of disease. According to previous understanding arthritis in the RS is the reactive one, which means that it is impossible to isolate its causative agent. However, there are the more and more authors suggesting that arthritis in the urogenital form of disease is caused by the infective agent in the affected joint. This suggestion is based on numerous studies on the presence of Chlmaydia trachomatis and Ureaplasma urealyticum in the inflamed joint by using new diagnostic methods in molecular biology published in the recent literature [1-3]. Besides, numerous studies of the humoral and cell-mediated immune response to "triggering" bacteria in the affected joint have supported previous suggestions [4-7]. Aim of the study was to determine whether synovial fluid T-cells specifically recognize the "triggering" bacteria presumably responsible for the Reiter's syndrome. METHOD The 3H-thymidine uptake procedure for measuring lymphocyte responses was applied to lymphocytes derived concurrently from synovial fluid (SF) and from peripheral blood (PB) [8]. Ureaplasma antigen and mitogen PHA stimulated lymphocytes in 24 RS patients (24 PB samples, 9 SF samples) and the results were compared with those found in 10 patients with rheumatoid arthritis (RA) (10 PB samples, 5 SF samples). Preparation of ureaplasma antigen. Ureaplasma was cultured on cell-free liquid medium [9]. Sample of 8 ml was heat-inactivated for 15 minutes at 601C and permanently stirred with magnetic mixer. The sample was centrifuged at 2000 x g for 40 minutes and than deposits carefully carried to other sterile glass tubes (Corex) and recentrifuged at 9000 x g for 30 minutes. The deposit was washed 3 times in sterile 0.9% NaCl, and final sediment was resuspended in 1.2 ml sterile 0.9% NaCl. Bacteriology: Chlamydia trachomatis was isolated by cell culture using cycloheximide-treated McCoy cells [10], while Ureaplasma urealyticum was identified according to its biochemical properties grown on cell-free liquid medium [9]. RESULTS Proliferative response of the PB lymphocytes to stimulation by mitogen and ureaplasma antigen did not differ between RS and RA patients. Also, there was no difference in proliferative response of SF lymphocytes to mitogen stimulation between RS and RA patients (Figure 1). However, proliferation of SF lymphocytes stimulated by ureaplasma antigen was significantly elevated in RS patients compared with the control group. This difference is statistically significant (p<0.05) (Figure 2). Difference in proliferative response of the PB and SF lymphocytes stimulated by the ureaplasma antigen was not found in RS patients. DISCUSSION It was found that SF lymphocytes of RS patients showed significantly elevated proliferative response to stimulation by the ureaplasma antigen compared with SF lymphocytes of the control group. There was no difference when the lymphocytes were stimulated by the mitogen. Our findings suggest that elevated proliferative response of lymphocytes is the sign of stimulation cell-mediated immunity to antigen present in inflamed joint. Hence, the main immune response to Ureaplasma is on the cell-mediated level in the affected joint. This confirms the earlier finding reported by Ford et all. who concluded that synovial rather than peripheral blood lymphocytes indicate the microbiological cause of arthritis [11,12]. Horowitz etal. demonstrated the correlation between clinical remission after antibiotic therapy and eradication of Ureaplasma, together with a decrease in cellular immune response synovial fluid lymphocytes to ureaplasma antigen stimulation [13]. In that study Horowitz did not find statisticaly significant difference of ureaplasma proliferative response between PB and SF lymphocytes in patients with RS. We obtained the same results. Than we concluded that sensibilization of immune system exist in the presence of foreign antigen in RS patients. The other authors demonstrated higher stimulation indices than the ones we found in our patients [11-15]. This difference may be the result of different preparation of antigens, in other words selection of serotype of Ureaplasma for antigen preparation different conditions of lymphocyte cultivation. We concluded that the presence of antigen, antigen-specific T cells and efficient antigen-presenting cells (CD4+T cells) in the joint of RS patients strongly suggests that a T-cell-mediated response to bacteria has the central role in the pathogenesis of Reiter's syndrom.
34

Netea, Mihai G. "Toward Identification of the Genetic Risk Profile for Cryptococcal Disease in HIV-Infected Patients." mBio 4, no. 5 (October 15, 2013). http://dx.doi.org/10.1128/mbio.00798-13.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
ABSTRACTCryptococcusspp. are important fungal pathogens that represent a major cause of morbidity and mortality in both immunocompetent and immunodeficient patients. Although cryptococcal disease is one of the major causes of death in HIV-infected patients, especially in sub-Saharan Africa, not all patients at risk with low CD4 counts develop the disease. It has been thus hypothesized that host genetic variation may represent an important susceptibility risk factor for this infection. In their recent study inmBio, Rohatgi et al. [S. Rohatgi et al., mBio 4(5):e00573-13, 2013, doi:10.1128/mBio.00573-13] present an important piece of evidence to support this hypothesis, by demonstrating that theFCGR3A158 F/V polymorphism has an important impact on susceptibility to cryptococcal disease in HIV-infected patients. The authors present both genetic evidence and immunological validation for the hypothesis that humoral immunity in general andFCGR3A-mediated uptake and antibody-dependent cellular cytotoxicity (ADCC) in particular play important roles in the pathogenesis ofCryptococcusinfection. Their discovery that the 158V allele of this polymorphism can increase the risk ofCryptococcusinfections up to 20-fold in homozygous individuals opens the possibility for risk stratification and personalized treatment of HIV-infected patients.
35

Furdychko, L. O. "Порушення імунного гомеостазу в ранній період розвитку виразкової хвороби шунка на тлі експериментальної пневмонії". Medical and Clinical Chemistry, № 1 (28 квітня 2017). http://dx.doi.org/10.11603/mcch.2410-681x.2017.v0.i1.7345.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Introduction. The article adduces the obtained results of the indices of immune homeostasis in the development of combined pathology (experimental pneumonia (EP) and gastric ulcer (GU) of the stomach). The immune system is characterized by the number of T-lymphocytes since they are the basis of the cellular component of the immuneresponse and the concentration of b-lymphocytes and circulating immune complexes (CIC), which are responsible for cellular immunity.The aim of the study – the content of T-lymphocytes, CEC to assess the immune system in the development of EP and GU.Materials and Methods. This experimental study was conducted on 39 guinea pigs-males. Experimental pneumonia caused by the method of V. N. Shliapnikova, T. L. Solodova [et al.] Gastric ulcer of the stomach wassimulated by the method of V. I. Komarova. We determined the content of T- and B-lymphocytes (SD3 and SD19) in the blood by the method of E. F. Chernushenko, L. S. Kohosova. The level of circulating immune complexes (CIC) in the blood by the method of Haskova V., Kaslik J..Results and Discussion. The results of experimental studies established that early period of development (4th and 8th day) of gastric ulcer and pneumonia is characterized by the following changes of immune homeostasis: decrease in the level of T-lymphocytes and elevated levels of lymphocytes and circulating immune complexes in the blood of Guinea pigs.Conclusion. A depression of cell growth and humoral types of immunity in terms of experimental pneumoniaand gastric ulcer was revealed.
36

"Correction for Fouts et al., Balance of cellular and humoral immunity determines the level of protection by HIV vaccines in rhesus macaque models of HIV infection." Proceedings of the National Academy of Sciences 112, no. 18 (April 16, 2015): E2413. http://dx.doi.org/10.1073/pnas.1506010112.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Tshilenge, Georges. "Immunogeno: Protective mechanism for Rift Valley fever in the Democratic Republic of Congo." Onderstepoort J Vet Res 79, no. 2 (June 26, 2012). http://dx.doi.org/10.4102/ojvr.v79i2.483.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Rift Valley fever (RVF) is an acute, fever causing viral disease that affects domestic animals and humans. In Democratic Republic of Congo (DRC), this pathology is not well documented. No epidemic of the RVF has not been reported but sera samples collected in six provinces surveyed from 2005 to 2006 revealed 14% of apparent prevalence and, high apparent prevalence (20%) of antibodies against RVF virus was reported in Katanga Province during the same survey; this serological evidence was associated with abortions cases in Cattle (Mulumba et al. 2009). Livestock immunisation is important for control of Rift Valley fever virus (RVFV) epidemics; however immunisation of susceptible domestic animals in endemic countries does not protect animals against the clinical disease but prevents the propagation of virus to human population through reduction of the amplification degree in host animals. The humoral immunity is sufficient for protection for animals as well as for humans. The infection caused by RVFV leads to neutralisation of the immunity of the animal (Barnard 1979).Various immunological studies have been made on the characterisation of immune response during RVFV epidemics but, until now several studies have been concentrated on the response of the innate immune particularly based on signal interferon system than the response of the adaptive immune and cell mediated humoral immune. The available information on the immune response related to RVFV does not seem to provide enough information on various mechanisms of the response immune system.The aim of the study is based on mechanism of immune response system including protective effect of immunisation against RVFV. In addition, epidemiological and molecular studies will be assessed. As a matter of fact, following studies will be conducted:• evaluation of the immunological protection against Rift Valley fever in vaccinated and non- vaccinated cattle using IgG and IgM ELISAs in Katanga Province• assessment of cellular response to Rift Valley fever disease in vaccinated and naturally infected cattle• molecular characterisation of RVFV strains circulating in vaccinated and non vaccinated cattle• assessment of protective effect related to vaccinal strains in cattle, using a longitudinal survey.The studies will be carried out Northern Katanga Province within two areas, one with historyof circulation of RVFV and other without history RVFV circulation.Whole blood, spleen, liver, lymph node will be collected as target tissues from cattle carcasses. In addition, goats and sheeps samples will be collected alongside from each area in order to clarify the disease situation. Serological tests based on the detection of Ig M and Ig G will be used. DIVA tests, LAMP, and IHC techniques will be used. Within previously vaccinated areas in the above mentioned areas and those that are not vaccinated, the collected samples will be analysed using RT-PCR/RT-LAMP.In vitro experimental studies systems will be carried out using animal PMBCs that will be infected with wild type of RVF virus as well as with vaccinal strains, such as clones 13 and MP12 to characterise various cell types such as CD4 T cells, CD8 T cells, B-cells, NK cells and, macrophages will be studied with regard to activation and apoptosis signals on various post – infection days, using flow cytometry. A pool of animals will be vaccinated with the Clone 13 and another with the MP12 to determine the traceability. The monitoring of the immune response will be done through the measurement of immunoglobulin G (Ig G) and immunoglobulin M (Ig M). RT-PCR, spectrophotometer or Facs methods will be used for the dosage of cells T CD4 + and Cell T CD8+.

До бібліографії