Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Sci ; 185(2): 119-127, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-34735018

RESUMO

COVID-19 (Coronavirus Disease 2019), the disease caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus-2) is an ongoing global public health emergency. As understanding of the health effects of COVID-19 has improved, companies and agencies worldwide have worked together to identify therapeutic approaches, fast-track clinical trials and pathways for emergency use, and approve therapies for patients. This work has resulted in therapies that not only improve survival, reduce time of hospitalization, and time to recovery, but also include preventative measures, such as vaccines. This manuscript discusses development programs for 3 products that are approved or authorized for emergency use at the time of writing: VEKLURY (remdesivir, direct-acting antiviral from Gilead Sciences, Inc.), REGEN-COV (casirivimab and imdevimab antibody cocktail from Regeneron Pharmaceuticals Inc.), and Comirnaty (Pfizer-BioNTech COVID-19 Vaccine [Pfizer, Inc.-BioNTech]), and perspectives from the U.S. Food and Drug Administration.


Assuntos
COVID-19 , Hepatite C Crônica , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes , Antivirais , Vacina BNT162 , Vacinas contra COVID-19 , Combinação de Medicamentos , Humanos , SARS-CoV-2
2.
Toxicol Mech Methods ; 26(4): 270-5, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27098016

RESUMO

Safety assessment evaluating the presence of impurities, residual materials, and contaminants in vaccines is a focus of current research. Thresholds of toxicological concern (TTCs) are mathematically modeled levels used for assessing the safety of many food and medication constituents. In this study, six algorithms are selected from the open-access ToxTree software program to derive a method for calculating TTCs for vaccine constituents: In Vivo Rodent Micronucleus assay/LD50, Benigni-Bossa/LD50, Cramer Extended/LD50, In Vivo Rodent Micronucleus assay/TDLo, Benigni-Bossa/TDLo, and the Cramer Extended/TDLo. Using an initial dataset (n = 197) taken from INCHEM, RepDose, RTECS, and TOXNET, the chemicals were divided into two families: "positive" - based on the presence of structures associated with adverse outcomes, or "negative" - no such structures or having structures that appear to be protective of health. The final validation indicated that the Benigni-Bossa/LD50 method is the most appropriate for calculating TTCs for vaccine constituents. Final TTCs were designated as 18.06 µg/person and 20.61 µg/person for the Benigni-Bossa/LD50 positive and negative structural families, respectively.


Assuntos
Contaminação de Medicamentos , Modelos Teóricos , Software , Toxicologia/métodos , Vacinas/química , Vacinas/toxicidade , Adjuvantes Farmacêuticos/química , Adjuvantes Farmacêuticos/toxicidade , Algoritmos , Dose Letal Mediana , Conservantes Farmacêuticos/química , Conservantes Farmacêuticos/toxicidade , Relação Quantitativa Estrutura-Atividade
3.
J Immunother Cancer ; 4: 6, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26885368

RESUMO

BACKGROUND: Lymphodepletion enhances adoptive T cell transfer (ACT) therapy by activating the innate immune system via microbes released from the radiation-injured gut. Microbial components, such as LPS, are key mediators of total body irradiation (TBI) enhancement, but our ability to strategically use these toll-like receptor (TLR) agonists to bolster the potency of T cell-based therapies for cancer remains elusive. Herein, we used TLR4 agonist LPS as a tool to address how and when to use TLR agonists to effectively improve cancer immunotherapy. METHODS: To determine the mechanisms of how innate immune activation via lymphodepletion potentiated antitumor T cell immunity, we utilized the pmel-1 melanoma mouse model. B16F10-bearing mice were preconditioned with 5Gy TBI and given a tripartite ACT therapy (consisting of transferred pmel-1 CD8(+) T cells, vaccination with fowlpox encoding gp100, and IL-2) along with TLR4 agonist LPS. The timing of LPS administration and the requirement of individual components of the tripartite therapy were evaluated based on tumor growth and the phenotype of recovered splenocytes by flow cytometry. We also evaluated the role of non-toxic and clinically used TLR4 and TLR9 agonists-monophosphoryl lipid A (MPL) and CpG Oligodeoxynucleotide (CpG ODN), respectively- for ACT therapy. RESULTS: Here we report that while exogenous administration of LPS was able to enhance adoptively transferred CD8(+) T cells' tumor destruction, LPS treatment alone did not replace individual components of the tripartite ACT regimen, or obviate TBI. Moreover, we found that sequentially administering LPS during or one day prior to ACT therapy compromised tumor regression. In contrast, administering LPS after ACT potentiated the antitumor effectiveness of the regimen, thereby supporting the expansion of transferred tumor-specific CD8(+) T cells over host CD4(+) T cells. We also found that non-toxic TLR agonists MPL and CpG potentiated the antitumor activity of infused CD8(+) T cells. Finally, TBI was no longer needed to regress tumors in mice who were depleted of host CD4(+) T cells, given a tripartite ACT regimen and then treated with low dose LPS. CONCLUSIONS: Collectively, our results identify how and when to administer TLR agonists to augment T cell-based immunotherapy in the absence or presence of host preconditioning for treatment of advanced malignancies. Our findings have clinical implications for the design of next generation immune-based therapies for patients with cancer.

4.
Proc Natl Acad Sci U S A ; 112(2): 476-81, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548153

RESUMO

Lymphodepleting regimens are used before adoptive immunotherapy to augment the antitumor efficacy of transferred T cells by removing endogenous homeostatic "cytokine sinks." These conditioning modalities, however, are often associated with severe toxicities. We found that microRNA-155 (miR-155) enabled tumor-specific CD8(+) T cells to mediate profound antitumor responses in lymphoreplete hosts that were not potentiated by immune-ablation. miR-155 enhanced T-cell responsiveness to limited amounts of homeostatic γc cytokines, resulting in delayed cellular contraction and sustained cytokine production. miR-155 restrained the expression of the inositol 5-phosphatase Ship1, an inhibitor of the serine-threonine protein kinase Akt, and multiple negative regulators of signal transducer and activator of transcription 5 (Stat5), including suppressor of cytokine signaling 1 (Socs1) and the protein tyrosine phosphatase Ptpn2. Expression of constitutively active Stat5a recapitulated the survival advantages conferred by miR-155, whereas constitutive Akt activation promoted sustained effector functions. Our results indicate that overexpression of miR-155 in tumor-specific T cells can be used to increase the effectiveness of adoptive immunotherapies in a cell-intrinsic manner without the need for life-threatening, lymphodepleting maneuvers.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Citocinas/biossíntese , Células HEK293 , Humanos , Imunoterapia Adotiva , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Antígeno gp100 de Melanoma/genética , Antígeno gp100 de Melanoma/imunologia
5.
J Immunother ; 33(1): 1-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19952961

RESUMO

Lymphodepletion before adoptive cell transfer (ACT)-based immunotherapies can enhance anti-tumor responses by augmenting innate immunity, by increasing access to homeostatic cytokines, and by depressing the numbers of regulatory T cells and myeloid-derived suppressor cells. Although it is clear that high-dose total body irradiation given together with hematopoietic stem cell (HSC) transplantation effectively enhances ACT, the relationship between the intensity of lymphodepletion and tumor treatment efficacy has not been systematically studied. Using the pmel-1 mouse model of self/tumor-reactive CD8 T cells, we observed a strong correlation between the intensity of the conditioning regimen and the efficacy of ACT-based treatments using linear regression analysis. This was the case for preparative total body irradiation administered either as a single dose (R=0.97, P<0.001) or in fractionated doses (R=0.94, P<0.001). Increased amounts of preparative total body irradiation were directly correlated with progressively more favorable ratios of transferred tumor-reactive CD8 T cells toward endogenous cells with the potential for inhibitory activity including: CD4 cells (potentially T regulatory cells); Gr1 cells (which are capable of functioning as myeloid-derived suppressor cells); and endogenous CD8 and natural killer 1.1 cells (that can act as "sinks" for homeostatic cytokines in the postablative setting). With increasing ablation, we also observed elevated lipopolysaccharide levels in the sera and heightened levels of systemic inflammatory cytokines. Thus, increased intensity lymphodepletion triggers enhanced tumor treatment efficacy and the benefits of high-dose total body irradiation must be titrated against its risks.


Assuntos
Imunoterapia/métodos , Depleção Linfocítica/métodos , Neoplasias/terapia , Subpopulações de Linfócitos T/transplante , Linfócitos T/transplante , Transferência Adotiva , Animais , Separação Celular , Citometria de Fluxo , Camundongos , Camundongos Transgênicos , Subpopulações de Linfócitos T/efeitos da radiação , Linfócitos T/efeitos da radiação , Irradiação Corporal Total
6.
Nat Med ; 15(7): 808-13, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525962

RESUMO

Self-renewing cell populations such as hematopoietic stem cells and memory B and T lymphocytes might be regulated by shared signaling pathways. The Wnt-beta-catenin pathway is an evolutionarily conserved pathway that promotes hematopoietic stem cell self-renewal and multipotency by limiting stem cell proliferation and differentiation, but its role in the generation and maintenance of memory T cells is unknown. We found that induction of Wnt-beta-catenin signaling by inhibitors of glycogen sythase kinase-3beta or the Wnt protein family member Wnt3a arrested CD8(+) T cell development into effector cells. By blocking T cell differentiation, Wnt signaling promoted the generation of CD44(low)CD62L(high)Sca-1(high)CD122(high)Bcl-2(high) self-renewing multipotent CD8(+) memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets. These findings reveal a key role for Wnt signaling in the maintenance of 'stemness' in mature memory CD8(+) T cells and have major implications for the design of new vaccination strategies and adoptive immunotherapies.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Memória Imunológica , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Fator 1-alfa Nuclear de Hepatócito , Receptores de Hialuronatos/análise , Selectina L/análise , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Fator 1 de Transcrição de Linfócitos T/fisiologia , beta Catenina/fisiologia
7.
Blood ; 114(3): 596-9, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19471017

RESUMO

Interleukin-17 (IL-17)-secreting CD8(+) T cells have been described, but they have not been thoroughly studied and they do not have a known role in cancer immunotherapy. We skewed CD8(+) T cells to secrete IL-17 through priming in Th17-polarizing conditions. IL-17-producing CD8(+) T cells demonstrated reduced expression of Eomes and diminished cytolytic differentiation in vitro. However, after adoptive transfer, these cells converted to interferon-gamma-producing effector cells and mediated regression of large, established tumors. This improved antitumor immunity was associated with increased expression of IL-7R-alpha, decreased expression of killer cell lectin-like receptor G1, and enhanced persistence of the transferred cells. This report is the first description of a cancer therapy with IL-17-secreting CD8(+) T cells. These findings have implications for the improvement of CD8(+) T cell-based adoptive immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Imunoterapia Adotiva/métodos , Interleucina-17/metabolismo , Neoplasias Experimentais/terapia , Animais , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Interferon gama/biossíntese , Interleucina-17/biossíntese , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Receptores de Interleucina-7/biossíntese , Receptores Semelhantes a Lectina de Células NK/biossíntese , Subpopulações de Linfócitos T , Linfócitos T Citotóxicos/imunologia
8.
Blood ; 112(12): 4746-54, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18799724

RESUMO

Graft-versus-tumor effects can be achieved after allogeneic bone marrow transplantation in patients with malignancies of the kidney or hematopoietic system but are often accompanied by severe graft-versus-host-disease (GVHD). We sought to maximize graft-versus-tumor while minimizing GVHD using tumor-specific allogeneic effector T cells rather than open-repertoire T cells. We transferred allogeneic CD8(+) pmel-1 or CD4(+) TRP-1 T cells specific for the melanoma-associated antigens, glycoprotein 100 (gp100) and tyrosinase-related protein-1 (TRP-1), respectively, into B16-melanoma-bearing mice. Mice receiving a preparative regimen of nonmyeloablating (5 Gy) total body irradiation experienced the rapid rejection of tumor-specific allogeneic lymphocytes with no impact on tumor growth. However, when mice were given more intense total body irradiation conditioning regimens combined with autologous bone marrow transplantation, adoptively transferred allogeneic tumor-specific T lymphocytes persisted at detectable levels for several weeks and mediated significant regression of large, vascularized tumors. We found that the risk of GVHD was low when tumor-specific T cells were transferred and significant toxicity was observed only when substantial numbers of open repertoire allogeneic naive T cells were mixed with the tumor-specific lymphocytes. Taken together, these data indicate that the use of tumor-specific allogeneic CD8(+) T cells or CD4(+) can result in significant antitumor effects in the absence of measurable GVHD.


Assuntos
Transferência Adotiva/métodos , Incompatibilidade de Grupos Sanguíneos/imunologia , Linfócitos do Interstício Tumoral/fisiologia , Linfócitos do Interstício Tumoral/transplante , Complexo Principal de Histocompatibilidade/fisiologia , Carga Tumoral , Animais , Incompatibilidade de Grupos Sanguíneos/genética , Células Cultivadas , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Tumor/genética , Efeito Enxerto vs Tumor/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Indução de Remissão/métodos , Transplante Homólogo/imunologia , Transplante Homólogo/fisiologia , Carga Tumoral/imunologia
9.
Proc Natl Acad Sci U S A ; 105(23): 8061-6, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18523011

RESUMO

Nonmutated tissue differentiation antigens expressed by tumors are attractive targets for cancer immunotherapy, but the consequences of a highly effective antitumor immune response on self-tissue have not been fully characterized. We found that the infusion of ex vivo expanded adoptively transferred melanoma/melanocyte-specific CD8+ T cells that mediated robust tumor killing also induced autoimmune destruction of melanocytes in the eye. This severe autoimmunity was associated with the up-regulation of MHC class I molecules in the eye and high levels of IFN-gamma derived from both adoptively transferred CD8+ T cells and host cells. Furthermore, ocular autoimmunity required the presence of the IFN-gamma receptor on target tissues. Data compiled from >200 eyes and tumors in 10 independently performed experiments revealed a highly significant correlation (P < 0.0001) between the efficacy of tumor immunotherapy and the severity of ocular autoimmunity. Administration of high doses of steroids locally mitigated ocular autoimmunity without impairing the antitumor effect. These findings have particular importance for immunotherapies directed against self-antigens and highlight the need for targeting unique tumor antigens not expressed in critical tissues.


Assuntos
Antígenos de Neoplasias/imunologia , Autoimunidade/imunologia , Olho/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Olho/patologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunoterapia , Camundongos , Neoplasias/patologia , Receptores de Interferon/imunologia , Regulação para Cima , Receptor de Interferon gama
10.
Blood ; 112(2): 362-73, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18354038

RESUMO

CD4+ T cells can differentiate into multiple effector subsets, but the potential roles of these subsets in anti-tumor immunity have not been fully explored. Seeking to study the impact of CD4+ T cell polarization on tumor rejection in a model mimicking human disease, we generated a new MHC class II-restricted, T-cell receptor (TCR) transgenic mouse model in which CD4+ T cells recognize a novel epitope in tyrosinase-related protein 1 (TRP-1), an antigen expressed by normal melanocytes and B16 murine melanoma. Cells could be robustly polarized into Th0, Th1, and Th17 subtypes in vitro, as evidenced by cytokine, chemokine, and adhesion molecule profiles and by surface markers, suggesting the potential for differential effector function in vivo. Contrary to the current view that Th1 cells are most important in tumor rejection, we found that Th17-polarized cells better mediated destruction of advanced B16 melanoma. Their therapeutic effect was critically dependent on interferon-gamma (IFN-gamma) production, whereas depletion of interleukin (IL)-17A and IL-23 had little impact. Taken together, these data indicate that the appropriate in vitro polarization of effector CD4+ T cells is decisive for successful tumor eradication. This principle should be considered in designing clinical trials involving adoptive transfer-based immunotherapy of human malignancies.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citotoxicidade Imunológica , Melanoma/imunologia , Animais , Linhagem Celular Tumoral , Interferon Tipo I/imunologia , Interferon gama , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Gravidez/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/imunologia
11.
Clin Cancer Res ; 13(18 Pt 1): 5280-9, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875756

RESUMO

Lymphodepletion with chemotherapeutic agents or total body irradiation (TBI) before adoptive transfer of tumor-specific T cells is a critical advancement in the treatment of patients with melanoma. More than 50% of patients that are refractory to other treatments experience an objective or curative response with this approach. Emerging data indicate that the key mechanisms underlying how TBI augments the functions of adoptively transferred T cells include (a) the depletion of regulatory T cells (T(reg)) and myeloid-derived suppressor cells that limit the function and proliferation of adoptively transferred cells; (b) the removal of immune cells that act as "sinks" for homeostatic cytokines, whose levels increase after lymphodepletion; and (c) the activation of the innate immune system via Toll-like receptor 4 signaling, which is engaged by microbial lipopolysaccharide that translocated across the radiation-injured gut. Here, we review these mechanisms and focus on the effect of Toll-like receptor agonists in adoptive immunotherapy. We also discuss alternate regimens to chemotherapy or TBI, which might be used to safely treat patients with advanced disease and promote tumor regression.


Assuntos
Imunoterapia Adotiva , Neoplasias/terapia , Linfócitos T/transplante , Receptores Toll-Like/agonistas , Animais , Humanos , Intestinos/imunologia , Depleção Linfocítica , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Linfócitos T/imunologia , Receptores Toll-Like/efeitos da radiação , Irradiação Corporal Total
12.
J Clin Invest ; 117(8): 2197-204, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17657310

RESUMO

Lymphodepletion with total body irradiation (TBI) increases the efficacy of adoptively transferred tumor-specific CD8(+) T cells by depleting inhibitory lymphocytes and increasing homeostatic cytokine levels. We found that TBI augmented the function of adoptively transferred CD8(+) T cells in mice genetically deficient in all lymphocytes, indicating the existence of another TBI mechanism of action. Additional investigation revealed commensal gut microflora in the mesenteric lymph nodes and elevated LPS levels in the sera of irradiated mice. These findings correlated with increased dendritic cell activation and heightened levels of systemic inflammatory cytokines. Reduction of host microflora using antibiotics, neutralization of serum LPS using polymyxin B, or removal of LPS signaling components using mice genetically deficient in CD14 and TLR4 reduced the beneficial effects of TBI on tumor regression. Conversely, administration of microbial ligand-containing serum or ultrapure LPS from irradiated animals to nonirradiated antibody-lymphodepleted mice enhanced CD8(+) T cell activation and improved tumor regression. Administration of ultrapure LPS to irradiated animals further enhanced the number and function of the adoptively transferred cells, leading to long-term cure of mice with large B16F10 tumors and enhanced autoimmune vitiligo. Thus, disruption of the homeostatic balance between the host and microbes can enhance cell-based tumor immunotherapy.


Assuntos
Transferência Adotiva , Translocação Bacteriana/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias Experimentais/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Irradiação Corporal Total , Animais , Antibacterianos/farmacologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/microbiologia , Doenças Autoimunes/patologia , Autoimunidade/efeitos dos fármacos , Autoimunidade/imunologia , Translocação Bacteriana/efeitos da radiação , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Citocinas/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Receptores de Lipopolissacarídeos/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Depleção Linfocítica , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/microbiologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Polimixina B/farmacologia , Transdução de Sinais/efeitos dos fármacos , Vitiligo/imunologia , Vitiligo/microbiologia , Vitiligo/patologia
13.
J Clin Invest ; 117(2): 492-501, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17273561

RESUMO

Depleting host immune elements with nonmyeloablative regimens prior to the adoptive transfer of tumor-specific CD8(+) T cells significantly enhances tumor treatment. In the current study, superior antitumor efficacy was achieved by further increasing the intensity of lymphodepletion to a level that required HSC transplantation. Surprisingly, the HSC transplant and not the increased lymphodepletion caused a robust expansion of adoptively transferred tumor-specific CD8(+) T cells. The HSC-driven cell expansion of effector, but not of naive, CD8(+) T cells was independent of in vivo restimulation by MHC class I-expressing APCs. Simultaneously, HSCs also facilitated the reconstitution of the host lymphoid compartment, including inhibitory elements, not merely via the production of progeny cells but by enhancing the expansion of cells that had survived lymphodepletion. Profound lymphodepletion, by myeloablation or by genetic means, focused the nonspecific HSC boost preferentially toward the transferred tumor-specific T cells, leading to successful tumor treatment. These findings indicate that CD8(+) T cell-mediated tumor responses can be efficiently driven by HSCs in the myeloablative setting and have substantial implications for the design of new antitumor immunotherapies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células-Tronco Hematopoéticas/imunologia , Melanoma Experimental/terapia , Animais , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunoterapia Adotiva , Depleção Linfocítica , Melanoma Experimental/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Condicionamento Pré-Transplante , Antígeno gp100 de Melanoma
14.
Nat Clin Pract Oncol ; 3(12): 668-81, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17139318

RESUMO

In a recent clinical trial involving patients with metastatic melanoma, immunosuppressive conditioning with fludarabine and cyclophosphamide resulted in a 50% response rate in robust long-term persistence of adoptively transferred T cells. Experimental findings indicate that lymphodepletion prior to adoptive transfer of tumor-specific T lymphocytes plays a key role in enhancing treatment efficacy by eliminating regulatory T cells and competing elements of the immune system ('cytokine sinks'). Newly emerging animal data suggest that more profound lymphoablative conditioning with autologous hematopoetic stem-cell rescue might further enhance treatment results. Here we review recent advances in adoptive immunotherapy of solid tumors and discuss the rationale for lymphodepleting conditioning. We also address safety issues associated with translating experimental animal results of total lymphoid ablation into clinical practice.


Assuntos
Imunoterapia Adotiva , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , Linfócitos do Interstício Tumoral
15.
J Exp Med ; 202(7): 907-12, 2005 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-16203864

RESUMO

Depletion of immune elements before adoptive cell transfer (ACT) can dramatically improve the antitumor efficacy of transferred CD8+ T cells, but the specific mechanisms that contribute to this enhanced immunity remain poorly defined. Elimination of CD4+CD25+ regulatory T (T reg) cells has been proposed as a key mechanism by which lymphodepletion augments ACT-based immunotherapy. We found that even in the genetic absence of T reg cells, a nonmyeloablative regimen substantially augmented CD8+ T cell reactivity to self-tissue and tumor. Surprisingly, enhanced antitumor efficacy and autoimmunity was caused by increased function rather than increased numbers of tumor-reactive T cells, as would be expected by homeostatic mechanisms. The gammaC cytokines IL-7 and IL-15 were required for augmenting T cell functionality and antitumor activity. Removal of gammaC cytokine-responsive endogenous cells using antibody or genetic means resulted in the enhanced antitumor responses similar to those seen after nonmyeloablative conditioning. These data indicate that lymphodepletion removes endogenous cellular elements that act as sinks for cytokines that are capable of augmenting the activity of self/tumor-reactive CD8+ T cells. Thus, the restricted availability of homeostatic cytokines can be a contributing factor to peripheral tolerance, as well as a limiting resource for the effectiveness of tumor-specific T cells.


Assuntos
Transferência Adotiva/métodos , Autoimunidade/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Linfopenia/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Irradiação Corporal Total
16.
J Clin Invest ; 115(6): 1616-26, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15931392

RESUMO

T cell differentiation is a progressive process characterized by phenotypic and functional changes. By transferring tumor-specific CD8+ T cells into tumor-bearing mice at various stages of differentiation, we evaluated their efficacy for adoptive immunotherapy. We found that administration of naive and early effector T cells, in combination with active immunization and IL-2, resulted in the eradication of large, established tumors. Despite enhanced in vitro antitumor properties, more-differentiated effector T cells were less effective for in vivo tumor treatment. Several events may underlie this paradoxical phenomenon: (a) downregulation of lymphoid-homing and costimulatory molecules; (b) inability to produce IL-2 and access homeostatic cytokines; and (c) entry into a proapoptotic and replicative senescent state. While the progressive acquisition of terminal effector properties is characterized by pronounced in vitro tumor killing, in vivo T cell activation, proliferation, and survival are progressively impaired. These findings suggest that the current methodology for selecting T cells for transfer is inadequate and provide new criteria for the generation and the screening of optimal lymphocyte populations for adoptive immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Apoptose/imunologia , Linfócitos T CD8-Positivos/transplante , Proliferação de Células , Sobrevivência Celular/imunologia , Senescência Celular/imunologia , Interleucina-2/biossíntese , Ativação Linfocitária/imunologia , Melanoma Experimental/terapia , Camundongos , Receptores de Retorno de Linfócitos/biossíntese , Subpopulações de Linfócitos T/transplante
17.
Curr Opin Immunol ; 17(2): 195-201, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15766681

RESUMO

Adoptive T-cell immunotherapy combined with non-myeloablative lymphodepletion has emerged as the most effective immunotherapy treatment for patients with metastatic melanoma (objective response rates of 50%). The mechanisms underlying this major advance in the field of immunotherapy include the elimination of regulatory elements and increased access to activating cytokines. This results in the activation of low-affinity T cells, enabling them to destroy tumors. We propose that a more complete depletion of the patient's immune system followed by transplantation with hematopoietic stem cells, which can be genetically modified, would be more effective in the treatment of metastatic cancer.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Imunoterapia Adotiva , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Proliferação de Células , Humanos , Ativação Linfocitária/imunologia , Linfopenia/imunologia , Linfopenia/terapia , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
18.
J Immunol ; 173(12): 7209-16, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15585842

RESUMO

It has been suggested that antitumor T cells specifically traffic to the tumor site, where they effect tumor destruction. To test whether tumor-reactive CD8(+) T cells specifically home to tumor, we assessed the trafficking of gp100-specific pmel-1 cells to large, vascularized tumors that express or do not express the target Ag. Activation of tumor-specific CD8(+) pmel-1 T cells with IL-2 and vaccination with an altered peptide ligand caused regression of gp100-positive tumors (B16), but not gp100-negative tumors (methylcholanthrene 205), implanted on opposing flanks of the same mouse. Surprisingly, we found approximately equal and very large numbers of pmel-1 T cells (>25% of all lymphocytes) infiltrating both Ag-positive and Ag-negative tumors. We also found evidence of massive infiltration and proliferation of activated antitumor pmel-1 cells in a variety of peripheral tissues, including lymph nodes, liver, spleen, and lungs, but not peripheral blood. Most importantly, evidence for T cell function, as measured by production of IFN-gamma, release of perforin, and activation of caspase-3 in target cells, was confined to Ag-expressing tumor. We thus conclude that CD8(+) T cell-mediated destruction of tumor is the result of specific T cell triggering at the tumor site. The ability to induce ubiquitous homing and specific tumor destruction may be important in the case of noninflammatory metastatic tumor foci.


Assuntos
Linfócitos T CD8-Positivos/transplante , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Movimento Celular/imunologia , Citotoxicidade Imunológica , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/transplante , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Citotoxicidade Imunológica/genética , Fibrossarcoma/patologia , Fibrossarcoma/prevenção & controle , Fibrossarcoma/terapia , Imunoterapia Adotiva/métodos , Ativação Linfocitária/genética , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/administração & dosagem , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Transplante de Neoplasias/imunologia , Transplante de Neoplasias/métodos , Transplante de Neoplasias/patologia , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Antígeno gp100 de Melanoma
19.
J Virol ; 77(6): 3851-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610161

RESUMO

Recent studies demonstrated the ability of the recombinant autonomous parvoviruses MVMp (fibrotropic variant of the minute virus of mice) and H-1 to transduce therapeutic genes in tumor cells. However, recombinant vector stocks are contaminated by replication-competent viruses (RCVs) generated during the production procedure. To reduce the levels of RCVs, chimeric recombinant vector genomes were designed by replacing the right-hand region of H-1 virus DNA with that of the closely related MVMp virus DNA and conversely. Recombinant H-1 and MVMp virus pseudotypes were also produced with this aim. In both cases, the levels of RCVs contaminating the virus stocks were considerably reduced (virus was not detected in pseudotyped virus stocks, even after two amplification steps), while the yields of vector viruses produced were not affected. H-1 virus could be distinguished from MVMp virus by its restriction in mouse cells at an early stage of infection prior to detectable viral DNA replication and gene expression. The analysis of the composite viruses showed that this restriction could be assigned to a specific genomic determinant(s). Unlike MVMp virus, H-1 virus capsids were found to be a major determinant of the greater permissiveness of various human cell lines for this virus.


Assuntos
Vetores Genéticos , Vírus Miúdo do Camundongo/genética , Parvoviridae/genética , Proteínas Recombinantes de Fusão/metabolismo , Recombinação Genética , Replicação Viral , Animais , Linhagem Celular , Engenharia Genética/métodos , Humanos , Camundongos , Vírus Miúdo do Camundongo/fisiologia , Parvoviridae/fisiologia , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...