Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Cell Dev Biol ; 11: 1225677, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37492225

RESUMO

Epithelial tissues are crucial to maintaining healthy organization and compartmentalization in various organs and act as a first line of defense against infection in barrier organs such as the skin, lungs and intestine. Disruption or injury to these barriers can lead to infiltration of resident or foreign microbes, initiating local inflammation. One often overlooked aspect of this response is local changes in tissue mechanics during inflammation. In this mini-review, we summarize known molecular mechanisms linking disruption of epithelial barrier function to mechanical changes in epithelial tissues. We consider direct mechanisms, such as changes in the secretion of extracellular matrix (ECM)-modulating enzymes by immune cells as well as indirect mechanisms including local activation of fibroblasts. We discuss how these mechanical changes can modulate local immune cell activity and inflammation and perturb epithelial homeostasis, further dysregulating epithelial barrier function. We propose that this two-way relationship between loss of barrier function and altered tissue mechanics can lead to a positive feedback loop that further perpetuates inflammation. We discuss this cycle in the context of several chronic inflammatory diseases, including inflammatory bowel disease (IBD), liver disease and cancer, and we present the modulation of tissue mechanics as a new framework for combating chronic inflammation.

3.
BioDrugs ; 37(1): 21-33, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36571696

RESUMO

The clinical approval of immune checkpoint inhibitors is an important advancement in the field of cancer immunotherapy. However, the percentage of beneficiaries is still limited and it is becoming clear that combination therapies are required to further enhance the treatment efficacy. The potential of strategies targeting the immunoregulatory network by "hitting the gas pedal" as opposed to "blocking the brakes" is being recognized and intensively investigated. Hence, next to immune checkpoint inhibitors, agonists of co-stimulatory receptors of the tumor necrosis factor superfamily (TNF-SF) are emerging as promising options to expand the immunomodulatory toolbox. In this review the development of different categories of recombinant antibody and ligand-based agonists of 4-1BB, OX40, and GITR is summarized and discussed in the context of the challenges presented by the structural and mechanistical features of the TNFR-SF. An overview of current formats, trends, and clinical studies is provided.


Assuntos
Neoplasias , Humanos , Inibidores de Checkpoint Imunológico , Fator de Necrose Tumoral alfa , Imunoterapia
4.
Blood ; 140(16): 1750-1751, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36264591
5.
Cancer Immunol Immunother ; 69(11): 2291-2303, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32504247

RESUMO

Target expression heterogeneity and the presence of an immunosuppressive microenvironment can hamper severely the efficiency of immunotherapeutic approaches. We have analyzed the potential to encounter and overcome such conditions by a combinatory two-target approach involving a bispecific antibody retargeting T cells to tumor cells and tumor-directed antibody-fusion proteins with costimulatory members of the B7 and TNF superfamily. Targeting the tumor-associated antigens EpCAM and EGFR with the bispecific antibody and costimulatory fusion proteins, respectively, we analyzed the impact of target expression and the influence of the immunosuppressive factors IDO, IL-10, TGF-ß, PD-1 and CTLA-4 on the targeting-mediated stimulation of T cells. Here, suboptimal activity of the bispecific antibody at diverse EpCAM expression levels could be effectively enhanced by targeting-mediated costimulation by B7.1, 4-1BBL and OX40L in a broad range of EGFR expression levels. Furthermore, the benefit of combined costimulation by B7.1/4-1BBL and 4-1BBL/OX40L was demonstrated. In addition, the expression of immunosuppressive factors was shown in all co-culture settings, where blocking of prominent factors led to synergistic effects with combined costimulation. Thus, targeting-mediated costimulation showed general promise for a broad application covering diverse target expression levels, with the option for further selective enhancement by the identification and blockade of main immunosuppressive factors of the particular tumor environment.


Assuntos
Anticorpos Biespecíficos/imunologia , Antígenos de Neoplasias/imunologia , Molécula de Adesão da Célula Epitelial/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Anticorpos Biespecíficos/farmacologia , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Evasão Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
6.
Mol Cancer Ther ; 18(7): 1278-1288, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31040163

RESUMO

IL15 and costimulatory receptors of the tumor necrosis superfamily (TNFRSF) have shown great potential to support and drive an antitumor immune response. However, their efficacy as monotherapy is limited. Here, we present the development of a novel format for a trifunctional antibody-fusion protein that combines and focuses the activity of IL15/TNFSF-ligand in a targeting-mediated manner to the tumor site. The previously reported format consisted of a tumor-directed antibody (scFv), IL15 linked to an IL15Rα-fragment (RD), and the extracellular domain of 4-1BBL, where noncovalent trimerization of 4-1BBL into its functional unit led to a homotrimeric molecule with 3 antibody and 3 IL15-RD units. To reduce the size and complexity of the molecule, we have now designed a second format, where 4-1BBL is introduced as single-chain (sc), that is 3 consecutively linked 4-1BBL ectodomains. Thus, a monomeric trifunctional fusion protein presenting only 1 functional unit of each component was generated. Interestingly, the in vitro activity on T-cell stimulation was conserved or even enhanced for the soluble and target-bound molecule, respectively. Also, in a lung tumor mouse model, comparable antitumor effects were observed. Furthermore, corroborating the concept, OX40L and GITRL were also successfully incorporated into the novel single-chain format and the advantage of target-bound trifunctional versus corresponding combined bifunctional fusion proteins demonstrated by measuring T-cell proliferation and cytotoxic potential in vitro and antitumor effects of RD_IL15_scFv_scGITRL in a lung tumor mouse model in vivo Thus, the trifunctional antibody-fusion protein single-chain format constitutes a promising innovative platform for further therapeutic developments.


Assuntos
Imunoterapia/métodos , Interleucina-15/imunologia , Neoplasias Pulmonares/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Proteínas Recombinantes de Fusão/farmacologia
7.
Methods Mol Biol ; 1827: 351-364, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30196506

RESUMO

Antibody-fusion proteins with ligands, e.g., of the TNF superfamily (TNFSF) can be adequately produced in mammalian expression systems. Here, we describe the transient production in adherent and suspension human embryonic kidney cells at laboratory scale, followed by purification procedures applying protein A and immobilized metal affinity chromatography for proteins with Fc domain and 6 × histidine-tag, respectively. In addition, characterization of the purified proteins by size exclusion chromatography is described.


Assuntos
Anticorpos/isolamento & purificação , Cromatografia de Afinidade/métodos , Cromatografia em Gel/métodos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Fatores de Necrose Tumoral/metabolismo , Células HEK293 , Histidina/metabolismo , Humanos , Ligantes , Oligopeptídeos/metabolismo
8.
Oncoimmunology ; 7(9): e1471442, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30228940

RESUMO

Co-stimulatory signals induced by ligands of the tumor necrosis factor superfamily (TNFSF) play a central role in T cell activation and have emerged as a promising strategy in cancer immunotherapy. Here, we established a novel class of bifunctional co-stimulatory fusion proteins with the aim to boost T cell activation at the level of T cell - antigen-presenting cell (APC) interaction. These novel dual-acting cytokine fusion proteins were created by connecting two different homotrimeric TNFSF ligands to form homotrimeric bifunctional molecules (Duokines) or by connecting single-chain derivatives of two different homotrimeric TNFSF with a single, flexible linker (single-chain Duokines, scDuokines). By linking the TNFSF ligands 4-1BBL, OX40L and CD27L in all possible combinations, cis-acting Duokines were generated that act on the same or adjacent T cells, while combining CD40L with 4-1BBL, OX40L and CD27L resulted in trans-acting Duokines acting simultaneously on APCs and T cells. In vitro, co-stimulation of T cells was seen for cis- and trans-acting Duokines and scDuokines in an antigen-independent as well as antigen-specific setting. Trans-acting molecules furthermore activated B cells, which represent a subclass of APCs. In a pilot experiment using the syngeneic B16-FAP mouse tumor model scDuokines displayed antitumoral activity in vivo in combination with a primary T cell-activating bispecific antibody, evident from reduced number of lung metastasis compared to the antibody-only treated group. Our data show that the bifunctional, co-stimulatory duokines are capable to enhance T cell-mediated anti-tumor immune responses, suggesting that they can serve as a new class of immuno-stimulatory molecules for use in cancer immunotherapy strategies.

9.
Oncoimmunology ; 6(12): e1361594, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29209565

RESUMO

Therapeutic strategies aiming for the induction of an effective immune response at the tumor site can be severely hampered by the encounter of an immunosuppressive microenvironment. We investigated here the potential of concerted costimulation by tumor-directed antibody-fusion proteins with B7.1, 4-1BBL and OX40L to enforce bispecific antibody-induced T cell stimulation in presence of recognized immunosuppressive factors including IL-10, TGF-ß, indoleamine 2,3-dioxygenase (IDO), PD-L1 and regulatory T cells. The expression and activity of these factors was demonstrated in the HT1080-FAP/PBMC co-culture setting, where individual and combined costimulation were still capable to enhance T cell stimulation, even though the general activation level was reduced. Additional blockade of TGF-ß or PD-1 resulted especially effective in further enhancing the degree of T cell activation. Here, best outcome was achieved by combined costimulation of targeted 4-1BBL and B7.1. Furthermore, their individual impact on the proliferation of naïve, memory and effector CD8+ and CD4+ T cell subsets, suggest the coverage of a comprehensive T cell response. Thus, our costimulatory antibody-fusion proteins show great potential to support T cell activation in adverse conditions dictated by the tumor microenvironment.

10.
Oncoimmunology ; 5(11): e1238540, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27999756

RESUMO

Co-stimulation via receptors of the tumor necrosis factor superfamily (TNFSF) emerges as promising strategy to support antitumor immune responses. Targeted strategies with antibody-fusion proteins composed of a tumor-directed antibody part and the extracellular domain of a co-stimulatory ligand of the TNFSF constitute an attractive option to focus the co-stimulatory activity to the tumor site. Since TNFSF members intrinsically form functional units of non-covalently linked homotrimers, the protein engineering of suitable antibody-fusion proteins is challenging. Aiming for molecules of simple and stable configuration, we used TNFSF ligands in a single-chain format (scTNFSF), i.e., three units of the ectodomain connected by polypeptide linkers, folding into an intramolecular trimer. By fusing tumor-directed scFv antibody fragments directed against EpCAM or FAP to co-stimulatory scTNFSF molecules (sc4-1BBL, scOX40L, scGITRL or scLIGHT), a set of monomeric scFv-scTNFSF fusion proteins was generated. In comparison to the scFv-TNFSF format, defined by intermolecular homotrimerization via the TNFSF part, scFv-scTNFSF showed equal or enhanced co-stimulatory activity despite reduced avidity in antibody binding. In addition, enhanced serum stability and improved bioavailability in mice were observed. We show that the scFv-scTNFSF format can be applied to various members of the TNFSF, presenting targeting-dependent co-stimulatory activity. Hence, this format exhibits favorable properties that make it a promising choice for further therapeutic fusion protein development.

11.
Pharmacol Ther ; 154: 57-66, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26145167

RESUMO

The potential of immunomodulatory proteins, in particular cytokines, for cancer therapy is well recognized, but hampered by the toxicity associated with their systemic application. In order to address this problem, targeted delivery by antibody fusion proteins has been early proposed and their development intensively pursued over the last decade. Here, factors influencing the selection and modification of cytokines and antibody formats for this approach are being discussed, indicating current developments and translational advances in the field.


Assuntos
Anticorpos Monoclonais/imunologia , Citocinas/imunologia , Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/imunologia , Humanos , Mediadores da Inflamação/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/tratamento farmacológico , Linfócitos T Citotóxicos/imunologia
12.
Cancer Res ; 74(18): 5256-65, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25035394

RESUMO

Deregulated molecular signaling pathways are responsible for the altered adhesive, migratory, and invasive properties of cancer cells. The different breast cancer subtypes are characterized by the expression of distinct miRNAs, short non-coding RNAs that posttranscriptionally modulate the expression of entire gene networks. Profiling studies have revealed downregulation of miR149 in basal breast cancer. Here, we show that miR149 expression severely impairs cell spreading, migration, and invasion of basal-like breast cancer cells. We identify signaling molecules, including the small GTPases Rap1a and Rap1b, downstream of integrin receptors as miR149 targets, providing an explanation for the defective Src and Rac activation during cell adhesion and spreading upon miR149 expression. Suppression of cell spreading by miR149 could be rescued, at least in part, by expression of constitutively active Rac. Finally, we demonstrate that increased miR149 levels block lung colonization in vivo. On the basis of our findings, we propose that miR149 downregulation in basal breast cancer facilitates the metastatic dissemination of tumor cells by supporting aberrant Rac activation. Cancer Res; 74(18); 5256-65. ©2014 AACR.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular/genética , MicroRNAs/genética , Animais , Neoplasias da Mama/metabolismo , Adesão Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Camundongos SCID , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
13.
Mol Cancer Ther ; 13(1): 112-21, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198185

RESUMO

Influencing the cytokine receptor network that modulates the immune response holds great potential for cancer immunotherapy. Although encouraging results have been obtained by focusing on individual members of the common γ-chain (γc) receptor family and TNF receptor superfamily so far, combination strategies might be required to further improve the effectiveness of the antitumor response. Here, we propose the combination of interleukin (IL)-15 and 4-1BBL in a single, tumor-directed molecule. Therefore, a trifunctional antibody fusion protein was generated, composed of a tumor-specific recombinant antibody, IL-15 linked to a fragment of the IL-15Rα chain (RD) and the extracellular domain of 4-1BBL. In soluble and targeted forms, the trifunctional antibody fusion protein RD_IL-15_scFv_4-1BBL was shown to stimulate activated T-cell proliferation and induce T-cell cytotoxicity to a similar degree as the bifunctional scFv_RD_IL-15 fusion protein. On the other hand, in targeted form, the trifunctional fusion protein was much more effective in inducing T-cell proliferation and IFN-γ release of unstimulated peripheral blood mononuclear cells (PBMC). Here, the additional signal enhancement could be attributed to the costimulatory activity of 4-1BBL, indicating a clear benefit for the simultaneous presentation of IL-15 and 4-1BBL in one molecule. Furthermore, the trifunctional antibody fusion protein was more effective than the corresponding bifunctional fusion proteins in reducing metastases in a tumor mouse model in vivo. Hence, the targeted combination of IL-15 and 4-BBL in the form of a trifunctional antibody-fusion protein is a promising new approach for cancer immunotherapy.


Assuntos
Ligante 4-1BB/imunologia , Imunoterapia , Interleucina-15/uso terapêutico , Neoplasias/terapia , Ligante 4-1BB/genética , Ligante 4-1BB/uso terapêutico , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Interleucina-15/genética , Interleucina-15/imunologia , Interleucina-15/urina , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/uso terapêutico , Linfócitos T Citotóxicos/imunologia
14.
BioDrugs ; 28(2): 123-31, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24018469

RESUMO

Treatment with cytokines holds great potential for cancer immunotherapy, but is generally restricted by systemic toxicity. Tumor-directed targeting in the form of antibody fusion proteins appears to be an attractive strategy to overcome this problem. In the last twenty years, continuous efforts in developing appropriate molecules have retrieved a variety of antibody fusion proteins that reveal promising therapeutic effects in preclinical studies. Currently, several candidates are in clinical evaluation. Here, recent developments exploring diverse antibody formats, tumor targets and cytokines of different families as well as strategies addressing cytokine modification or presentation are discussed and clinical trials summarized at a glance. Thus, antibody-cytokine fusion proteins are becoming progressively improving immunologic reagents that raise expectations mainly for combinatorial cancer therapies.


Assuntos
Citocinas/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Anticorpos/uso terapêutico , Citocinas/administração & dosagem , Citocinas/imunologia , Humanos , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
15.
Cancer Immunol Immunother ; 62(8): 1369-80, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23715927

RESUMO

Combinatory strategies are becoming of increasing interest in cancer immunotherapy. Costimulation by individual members of the immunoglobulin-like (Ig)- and TNF superfamily have already shown promising antitumor potential, thus prompting the exploration of their synergistic abilities in combinatorial approaches. Here, we pursued a targeted strategy with antibody-fusion proteins composed of a tumor-directed antibody and the extracellular domain of the costimulatory ligand B7.1, 4-1BBL, OX40L, GITRL or LIGHT, respectively. Costimulatory activity was assessed in an experimental setting where initial T cell activation was induced by a bispecific antibody (tumor-related antigen × CD3). Advantage of combined targeted costimulation was shown for either B7.1 or 4-1BBL with OX40L, GITRL, LIGHT and 4-1BBL in terms of T cell proliferation and IFN-γ release. Since encouraging results were obtained by the combination of B7.1 and 4-1BBL, we adapted the model system for a time-shift setting. Here, enhanced proliferation and granzyme B expression as well as reduced PD-1 expression on the T cell population demonstrated the benefit of costimulation-assisted restimulation. Finally, the antitumor potential of this combinatorial setting was confirmed in vivo in a lung metastasis mouse model. Thus, combinatorial approaches with costimulatory antibody-ligand fusion proteins seem a promising strategy to be further investigated for cancer immunotherapy.


Assuntos
Imunoterapia/métodos , Neoplasias Experimentais/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Ligante 4-1BB/genética , Ligante 4-1BB/imunologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Granzimas/imunologia , Granzimas/metabolismo , Células HEK293 , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Ligantes , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/imunologia
16.
Oncoimmunology ; 1(7): 1213-1214, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23170284

RESUMO

Under physiological conditions, the trans-presentation of interleukin-15 (IL-15) by the IL-15 receptor α on the cell surface allows to confine and tune the IL-15-mediated immune responses. Therefore, targeting strategies that mimic this situation at the tumor sites appear especially promising for anticancer immunotherapy.

17.
J Immunother ; 35(5): 418-29, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22576347

RESUMO

Initiation of a tumor-directed immune response and appropriate modulation of its progress are key issues in cancer immunotherapy. Combinatorial strategies addressing both aspects might therefore be especially suitable. Here, we report a targeted approach combining a bispecific antibody with 2 costimulatory antibody-ligand fusion proteins. According to the concept, the bispecific antibody (scDbFAP×CD3) retargets T cells in a MHC-independent manner to tumor cells, providing an artificial first signal that allows the costimulatory antibody-ligand fusion proteins (B7.2-Db and scFv-4-1BBL) likewise targeted to the tumor cells to modulate the T-cell response. In our model system, the target cells coexpress the fibroblast activation protein (FAP) and endoglin as antigens. ScDbFAPCD3 and B7.2-Db are targeted to FAP although by different antibody moieties, whereas scFv-4-1BBL is directed against endoglin. ScDbFAPCD3-induced T-cell stimulation could be enhanced by the addition of either B7.2-Db or scFv-4-1BBL and even further by the combination of both as shown in terms of cytokine release (interleukin-2/interferon γ), proliferation and activation marker expression (CD25). By combined costimulation, overall T-cell population strongly increased in activation-experienced memory phenotype accompanied by a decrease in naive phenotype. ScFv-4-1BBL-mediated costimulation of naive CD8+ T cells promoted the expansion and development of cytotoxic T cells with strong effector potential. Thus, combining a bispecific antibody with antibody-ligand fusion protein-mediated CD28 and 4-1BB costimulation in a targeted approach shows great potential to generate and shape an immune response at the tumor site. Therefore, the adaptation of this approach to other immune modulatory ligands and tumor-relevant targets seems to be promising.


Assuntos
Anticorpos Biespecíficos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos Biespecíficos/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Endoglina , Endopeptidases , Gelatinases/genética , Gelatinases/imunologia , Células HEK293 , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Subunidade alfa de Receptor de Interleucina-2/imunologia , Ligantes , Ativação Linfocitária , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Terapia de Alvo Molecular , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Transfecção
18.
Mol Cancer Ther ; 11(6): 1279-88, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22491823

RESUMO

Cytokines driving the immune response are powerful tools for cancer immunotherapy, but their application is generally limited by severe systemic toxicity. Targeted approaches by means of antibody-cytokine fusion proteins might enable focus on the cytokine activity to the tumor site, thereby reducing unwanted side effects. Here, we investigated the possibility to improve the efficiency of interleukin (IL)-15 presentation in a targeted approach by the incorporation of an IL-15Rα chain fragment, mimicking physiologic trans-presentation. Therefore, an antibody cytokine fusion protein (scFv_RD_IL-15) composed of an antibody moiety targeting the tumor stromal fibroblast activation protein (FAP), an extended IL-15Rαsushi domain (RD) and IL-15 was generated, exhibiting antibody-mediated specific binding and cytokine activity in soluble and targeted form. Comparative analysis with a corresponding antibody fusion protein devoid of RD (scFv_IL-15) showed for scFv_RD_IL-15 in solution enhanced stimulatory activity on Mo7e (IL-15Rßγ) cells and reduced proliferation response on CTLL-2 (IL-15Rαßγ) cells, while in FAP-targeted, that is, membrane-bound form, comparable proliferation of CTLL-2 (IL-15Rαßγ) cells was obtained. In addition, scFv_RD_IL-15 achieved in its soluble and target-bound form stronger proliferation and cytotoxicity on unstimulated and activated T cells, respectively. Furthermore, in vivo analysis in a lung metastasis tumor mouse model revealed a superior antitumor effect for scFv_RD_IL-15 in comparison with that obtained by an untargeted or RD missing version of IL-15 fusion protein. Thus, tumor-directed trans-presentation of IL-15 in association with RD in form of an antibody fusion protein seems to be a promising approach to further improve the antitumor effect of IL-15.


Assuntos
Antineoplásicos/farmacologia , Subunidade alfa de Receptor de Interleucina-15/genética , Interleucina-15/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Cadeia Única/genética , Animais , Apresentação de Antígeno , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Proliferação de Células , Endopeptidases , Gelatinases/imunologia , Gelatinases/metabolismo , Humanos , Imunoterapia , Interleucina-15/metabolismo , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mimetismo Molecular , Terapia de Alvo Molecular , Transplante de Neoplasias , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
19.
J Immunother ; 33(9): 925-34, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20948444

RESUMO

Activation of murine glucocorticoid-induced tumor necrosis factor-related receptor (mGITR) by its natural ligand (GITRL) or antiGITR agonist mAb enhances T-cell responses, inhibits regulatory T-cell (Treg)-mediated suppression and induces tumor immunity in a variety of murine tumor models. However, systemic administration of these costimulatory agents can lead to global T-cell activation and autoimmunity. To specifically manipulate the T-cell compartment in the tumor microenvironment we propose to target the tumor infiltrating T cells with a bispecific mGITRL fusion protein. For that purpose, mGITRL is linked to a single-chain antibody targeting fibroblast activation protein (FAP) as FAP expression is restricted to cancer-associated fibroblasts (CAFs) found in the stroma of epithelial cancers. AntiFAP-mGITRL fusion protein forms dimers and binds to murine GITR with 1.2 µM affinity and to murine FAP with 4.5 nM. The construct is able to costimulate CD8+ and CD4+ effector T cells resulting in increased proliferation, IFN-γ and IL-2 production. This costimulatory effect is enhanced when the fusion protein is bound to a FAP-positive cell line mimicking FAP CAFs. In suppression assays, membrane-bound antiFAP-mGITRL is 100-fold more effective in overcoming Treg-mediated suppression than unbound fusion protein. These studies suggest that targeted tumor therapy with antiFAP-mGITRL fusion protein could induce tumor rejection while minimizing autoimmune side effects.


Assuntos
Antígenos de Neoplasias/metabolismo , Imunoterapia , Neoplasias Experimentais/imunologia , Receptores de Fator de Crescimento Neural/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Endopeptidases , Gelatinases/genética , Gelatinases/imunologia , Gelatinases/metabolismo , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Ativação Linfocitária/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/tratamento farmacológico , Estrutura Terciária de Proteína/genética , Receptores de Fator de Crescimento Neural/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia
20.
Protein Eng Des Sel ; 23(11): 827-34, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20817756

RESUMO

Fusion of small recombinant antibody fragments to an albumin-binding domain (ABD) from streptococcal protein G strongly extends their plasma half-life. This ABD binds with nanomolar affinity to human (HSA) and mouse serum albumin (MSA). It was speculated that an increase in albumin-binding affinity should lead to a further increase in half-life. In the present study, we analyzed the effects of affinity and valency of the ABD on the pharmacokinetic properties of a bispecific single-chain diabody (scDb), applied previously to investigate various half-life extension strategies. The scDb is directed against carcinoembryonic antigen (CEA) and CD3 capable of mediating T cell retargeting to tumor cells. Two scDb derivatives with increased (scDb-ABD-H) and decreased (scDb-ABD-L) affinity as well as an scDb molecule fused to two ABD (scDb-ABD(2)) were generated and produced in mammalian cells. The altered binding of these constructs to HSA and MSA was confirmed by ELISA and quartz crystal microbalance measurements. All constructs bound efficiently to CEA and CD3-positive cells and were able to activate T cells in a target cell-dependent manner, although T cell activation was reduced in the presence of serum albumin. All three derivatives showed a strongly increased half-life in mice as compared with scDb. Compared with the wild-type scDb-ABD, the half-life of scDb-ABD-H exhibited a prolonged half-life and scDb-ABD-L a reduced half-life, while the half-life scDb-ABD(2) was almost identical to that of scDb-ABD. However, these changes were only moderate, indicating that the half-life-extending property of the ABD in mice is only weakly influenced by affinity for serum albumin or valency of albumin binding.


Assuntos
Anticorpos Biespecíficos/metabolismo , Anticorpos Biespecíficos/farmacocinética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Albumina Sérica/metabolismo , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Complexo CD3/imunologia , Antígeno Carcinoembrionário/imunologia , Linhagem Celular , Meia-Vida , Humanos , Interleucina-2/imunologia , Camundongos , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...