Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Toxicol Appl Pharmacol ; 487: 116961, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38740095

RESUMO

LEAD-452 is a humanized bispecific EGFR-targeted 4-1BB-agonistic trimerbody with a unique trimeric configuration compared to other 4-1BB-specific antibodies that are currently in development. Indeed, enhanced tumor-specific costimulation and very remarkable safety and efficacy profiles have been observed in mouse models. Here, we conducted for the first time a preclinical pharmacokinetic and toxicity study in non-human primates (NHP) (Macaca fascicularis). LEAD-452 exhibits comparable binding affinity for human and macaque targets, indicating its pharmacological significance for safety testing across species. The NHP were administered LEAD-452 in a series of ascending doses, ranging from 0.1 mg/kg to 10 mg/kg, and repeated doses up to 20 mg/kg. The administration of LEAD-452 was found to be clinically well tolerated, with no major related adverse effects observed. Furthermore, there have been no reported cases of liver toxicity, thrombocytopenia, and neutropenia, which are commonly associated with treatments using conventional anti-4-1BB IgG-based antibodies. In addition, neither IgM nor IgG-based anti-drug antibodies were detected in serum samples from NHP during the study, regardless of the dose of LEAD-452 administered. These results support the clinical development of LEAD-452 for the treatment of solid tumors.


Assuntos
Receptores ErbB , Macaca fascicularis , Animais , Receptores ErbB/imunologia , Humanos , Masculino , Feminino , Anticorpos Biespecíficos/farmacocinética , Anticorpos Biespecíficos/efeitos adversos , Relação Dose-Resposta a Droga
2.
Trends Immunol ; 40(3): 243-257, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30827461

RESUMO

The redirection of T cell activity towards cancer cells via targeting of tumor-associated antigens (TAAs) by soluble bispecific antibodies (bsAbs) or membrane-anchored chimeric antigen receptors is one of the most promising cancer immunotherapy strategies currently in development. We review here an emerging approach that combines aspects of antibody- and cell-based therapies: STAb immunotherapy, based on the endogenous secretion of T cell-redirecting bsAbs (STAb). STAb immunotherapies use ex vivo or in vivo genetic modifications of different cell types with nucleic acids or viral vectors encoding bsAbs; these can result in effective and persistent concentrations of antibodies. After introducing core concepts, we discuss plausible ways by which STAb strategies might be further developed to improve their potential efficacy and safety in preclinical and clinical testing.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Linfócitos B/fisiologia , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/imunologia , Engenharia Genética , Vetores Genéticos , Humanos , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante
3.
Mol Pharm ; 16(3): 1025-1035, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30726099

RESUMO

Monoclonal antibodies (mAbs) are currently used as therapeutic agents in different types of cancer. However, mAbs and antibody fragments developed so far show suboptimal properties in terms of circulation time and tumor penetration/retention. Here, we report the radiolabeling, pharmacokinetic evaluation, and determination of tumor targeting capacity of the previously validated anti-CEA MFE23-scFv-based N-terminal trimerbody (MFE23N-trimerbody), and the results are compared to those obtained for the monomeric MFE23-scFv. Dissection and gamma-counting studies performed with the 131I-labeled protein scaffolds in normal mice showed slower blood clearance for the trimerbody, and accumulation in the kidneys, the spleen, and the liver for both species. These, together with a progressive uptake in the small intestine, confirm a combined elimination scheme with hepatobiliary and urinary excretion. Positron emission tomography studies performed in a xenograft mouse model of human gastric adenocarcinoma, generated by subcutaneous administration of CEA-positive human MKN45 cells, showed higher tumor accumulation and tumor-to-muscle (T/M) ratios for 124I-labeled MFE23N-trimerbody than for MFE23-scFv. Specific uptake was not detected with PET imaging in CEA negative xenografts as indicated by low T/M ratios. Our data suggest that engineered intermediate-sized trivalent antibody fragments could be promising candidates for targeted therapy and imaging of CEA-positive tumors.


Assuntos
Antígeno Carcinoembrionário/imunologia , Tomografia por Emissão de Pósitrons/métodos , Anticorpos de Cadeia Única/farmacocinética , Neoplasias Gástricas/diagnóstico por imagem , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Radioisótopos do Iodo , Marcação por Isótopo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia de Alvo Molecular , Neoplasias Gástricas/tratamento farmacológico , Distribuição Tecidual , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Immunol Immunother ; 67(8): 1251-1260, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29869168

RESUMO

The recruitment of T-cells by bispecific antibodies secreted from adoptively transferred, gene-modified autologous cells has shown satisfactory results in preclinical cancer models. Even so, the approach's translation into the clinic will require incremental improvements to its efficacy and reduction of its toxicity. Here, we characterized a tandem T-cell recruiting bispecific antibody intended to benefit gene-based immunotherapy approaches, which we call the light T-cell engager (LiTE), consisting of an EGFR-specific single-domain VHH antibody fused to a CD3-specific scFv. We generated two LiTEs with the anti-EGFR VHH and the anti-CD3 scFv arranged in both possible orders. Both constructs were well expressed in mammalian cells as highly homogenous monomers in solution with molecular weights of 43 and 41 kDa, respectively. In situ secreted LiTEs bound the cognate antigens of both parental antibodies and triggered the specific cytolysis of EGFR-expressing cancer cells without inducing T-cell activation and cytotoxicity spontaneously or against EGFR-negative cells. Light T-cell engagers are, therefore, suitable for future applications in gene-based immunotherapy approaches.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Complexo CD3/imunologia , Receptores ErbB/imunologia , Imunoterapia , Neoplasias/terapia , Anticorpos de Cadeia Única/uso terapêutico , Linfócitos T/imunologia , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Ativação Linfocitária , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Células Tumorais Cultivadas
5.
J Cell Mol Med ; 20(5): 980-6, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26915562

RESUMO

We have recently described the response of human brain pericytes to lipopolysaccharide (LPS) through toll-like receptor 4 (TLR4). However, Gram-negative pathogen-associated molecular patterns include not only LPS but also peptidoglycan (PGN). Given that the presence of co-purified PGN in the LPS preparation previously used could not be ruled out, we decided to analyse the expression of the intracellular PGN receptors NOD1 and NOD2 in HBP and compare the responses to their cognate agonists and ultrapure LPS. Our findings show for the first time that NOD1 is expressed in pericytes, whereas NOD2 expression is barely detectable. The NOD1 agonist C12-iE-DAP induced IL6 and IL8 gene expression by pericytes as well as release of cytokines into culture supernatant. Moreover, we demonstrated the synergistic effects of NOD1 and TLR4 agonists on the induction of IL8. Using NOD1 silencing in HBP, we showed a requirement for C12-iE-DAP-dependent signalling. Finally, we could discriminate NOD1 and TLR4 pathways in pericytes by pharmacological targeting of RIPK2, a kinase involved in NOD1 but not in TLR4 signalling cascade. p38 MAPK and NF-κB appear to be downstream mediators in the NOD1 pathway. In summary, these results indicate that pericytes can sense Gram-negative bacterial products by both NOD1 and TLR4 receptors, acting through distinct pathways. This provides new insight about how brain pericytes participate in the inflammatory response and may have implications for disease management.


Assuntos
Lipopolissacarídeos/farmacologia , Proteína Adaptadora de Sinalização NOD1/genética , Peptidoglicano/farmacologia , Pericitos/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/metabolismo , Artérias Cerebrais/citologia , Artérias Cerebrais/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Interleucina-8/biossíntese , Interleucina-8/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/deficiência , Proteína Adaptadora de Sinalização NOD2/genética , Pericitos/citologia , Pericitos/metabolismo , Cultura Primária de Células , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23918357

RESUMO

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Assuntos
Anticorpos Antineoplásicos/imunologia , Biomarcadores Tumorais/imunologia , Imunoterapia/métodos , Proteínas Musculares/metabolismo , Neoplasias da Próstata/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Anticorpos Antineoplásicos/metabolismo , Especificidade de Anticorpos , Western Blotting , Técnicas de Visualização da Superfície Celular , Cromatografia de Afinidade , Cromatografia Líquida , Sistemas de Liberação de Medicamentos/métodos , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/terapia , Estatísticas não Paramétricas , Espectrometria de Massas em Tandem
7.
J Biol Chem ; 289(4): 2457-68, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24307174

RESUMO

Pericytes and mesenchymal stem cells (MSCs) are ontogenically related, and in fact, no significant phenotypic differences could be observed by flow cytometry. Transcriptome analysis of human pericytes and MSCs revealed that 43 genes were up-regulated more than 10-fold in pericytes compared with MSCs. Identification of Toll-like receptor 4 (TLR4) as one of the most abundant RNA species in pericytes with respect to MSCs and confirmation of TLR4 expression on the cell surface led us to obtain a comprehensive overview of the expression program of lipopolysaccharide (LPS)-stimulated pericytes. Transcriptional profiling of LPS-treated cells revealed that 22 genes were up-regulated more than 5-fold. Of them, 10 genes encoded chemokines and cytokines (CXCL10, CCL20, IL8, CXCL1, IL6, CCL2, IL1B, CXCL2, IL1A, and CXCL6), and three genes encoded adhesion molecules (ICAM1, VCAM1, and SELE). LPS induced nuclear translocation of the transcription factor NF-κB in stimulated pericytes. Moreover, inhibition of NF-κB activation by SC-514 blocked LPS-induced up-regulation of a subset of chemokine genes, confirming the key role of NF-κB in LPS signaling in pericytes. At the protein level, we assessed the secretion of the proinflammatory cytokines and chemokines IL-6, IL-8, CXCL1, CXCL2, CXCL3, and CCL2 not only after LPS treatment but also in HMGB1-stimulated pericytes. Up-regulation of the adhesion molecules ICAM-1 and VCAM-1 resulted in an increased adhesion of peripheral blood leukocytes to an LPS-treated pericyte monolayer. The role of pericytes in the inflammatory context has been scarcely addressed; according to these results, pericytes should be considered as active players in the inflammatory cascade with potential physiopathological implications.


Assuntos
Moléculas de Adesão Celular/biossíntese , Citocinas/biossíntese , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Pericitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/agonistas , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Moléculas de Adesão Celular/imunologia , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/metabolismo , NF-kappa B/imunologia , Pericitos/citologia , Pericitos/imunologia , Transdução de Sinais/imunologia , Tiofenos/farmacologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
8.
Microb Cell Fact ; 13: 116, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25112455

RESUMO

BACKGROUND: Recombinant antibodies are highly successful in many different pathological conditions and currently enjoy overwhelming recognition of their potential. There are a wide variety of protein expression systems available, but almost all therapeutic antibodies are produced in mammalian cell lines, which mimic human glycosylation. The production of clinical-grade antibodies in mammalian cells is, however, extremely expensive. Compared to mammalian systems, protein production in yeast strains such as Pichia pastoris, is simpler, faster and usually results in higher yields. RESULTS: In this work, a trivalent single-chain fragment variable (scFv)-based N-terminal trimerbody, specific for the human carcinoembryonic antigen (CEA), was expressed in human embryonic kidney 293 cells and in Pichia pastoris. Mammalian- and yeast-produced anti-CEA trimerbody molecules display similar functional and structural properties, yet, the yield of trimerbody expressed in P. pastoris is about 20-fold higher than in human cells. CONCLUSIONS: P. pastoris is an efficient expression system for multivalent trimerbody molecules, suitable for their commercial production.


Assuntos
Biotecnologia/métodos , Pichia/metabolismo , Anticorpos de Cadeia Única/biossíntese , Anticorpos de Cadeia Única/química , Antígeno Carcinoembrionário/imunologia , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Células HEK293 , Humanos , Proteínas Imobilizadas/metabolismo , Estabilidade Proteica , Estrutura Terciária de Proteína , Soro/metabolismo , Anticorpos de Cadeia Única/isolamento & purificação
9.
Mol Ther ; 21(2): 348-57, 2013 02.
Artigo em Inglês | MEDLINE | ID: mdl-23011032

RESUMO

Injection of oncolytic vesicular stomatitis virus (VSV) into established B16ova melanomas results in tumor regression, in large part by inducing innate immune reactivity against the viral infection, mediated by MyD88- and type III interferon (IFN)-, but not TLR-4-, signaling. We show here that intratumoral (IT) treatment with lipopolysaccharide (LPS), a TLR-4 agonist, significantly enhanced the local therapy induced by VSV by combining activation of different innate immune pathways. Therapy was further enhanced by co-recruiting a potent antitumor, adaptive T-cell response by using a VSV engineered to express the ovalbumin tumor-associated antigen ova, in combination with LPS. However, the combination of IT LPS with systemically delivered VSV resulted in rapid morbidity and mortality in the majority of mice. Decreasing the intravenous (IV) dose of VSV to levels at which toxicity was ameliorated did not enhance therapy compared with IT LPS alone. Toxicity of the systemic VSV + IT LPS regimen was associated with rapidly elevated levels of serum tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, which neither systemic VSV, nor IT LPS, alone induced. These data show that therapy associated with direct IT injections of oncolytic viruses can be significantly enhanced by combination with agonists of innate immune activation pathways, which are not themselves activated by the virus alone. Importantly, they also highlight possible, unforeseen dangers of combination therapies in which an immunotherapy, even delivered locally at the tumor site, may systemically sensitize the patient to a cytokine shock-like response triggered by IV delivery of oncolytic virus.


Assuntos
Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Receptor 4 Toll-Like/agonistas , Vesiculovirus/genética , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Terapia Genética , Vetores Genéticos , Imunidade Inata , Imunoterapia , Interferon-alfa/metabolismo , Interleucina-6/sangue , Interleucina-6/imunologia , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vírus Oncolíticos/genética , Transdução de Sinais , Linfócitos T/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Vesiculovirus/imunologia
10.
Oncoimmunology ; 13(1): 2338558, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38623463

RESUMO

T cell-based immunotherapies for solid tumors have not achieved the clinical success observed in hematological malignancies, partially due to the immunosuppressive effect promoted by the tumor microenvironment, where PD-L1 and TGF-ß play a pivotal role. However, durable responses to immune checkpoint inhibitors remain limited to a minority of patients, while TGF-ß inhibitors have not reached the market yet. Here, we describe a bispecific antibody for dual blockade of PD-L1 and TFG-ß, termed AxF (scFv)2, under the premise that combination with T cell redirecting strategies would improve clinical benefit. The AxF (scFv)2 antibody was well expressed in mammalian and yeast cells, bound both targets and inhibited dose-dependently the corresponding signaling pathways in luminescence-based cellular reporter systems. Moreover, combined treatment with trispecific T-cell engagers (TriTE) or CAR-T cells significantly boosted T cell activation status and cytotoxic response in breast, lung and colorectal (CRC) cancer models. Importantly, the combination of an EpCAMxCD3×EGFR TriTE with the AxF (scFv)2 delayed CRC tumor growth in vivo and significantly enhanced survival compared to monotherapy with the trispecific antibody. In summary, we demonstrated the feasibility of concomitant blockade of PD-L1 and TGF-ß by a single molecule, as well as its therapeutic potential in combination with different T cell redirecting agents to overcome tumor microenvironment-mediated immunosuppression.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Neoplasias Colorretais , Animais , Humanos , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/farmacologia , Antígeno B7-H1 , Neoplasias Colorretais/tratamento farmacológico , Linfócitos T , Fator de Crescimento Transformador beta , Microambiente Tumoral
11.
Antibodies (Basel) ; 13(2)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38804302

RESUMO

Immune checkpoint blockade has changed the treatment paradigm for advanced solid tumors, but the overall response rates are still limited. The combination of checkpoint blockade with anti-4-1BB antibodies to stimulate tumor-infiltrating T cells has shown anti-tumor activity in human trials. However, the further clinical development of these antibodies has been hampered by significant off-tumor toxicities. Here, we generated an anti-4-1BB/EGFR/PD-L1 trispecific antibody consisting of a triple-targeting tandem trimerbody (TT) fused to an engineered silent Fc region. This antibody (IgTT-4E1-S) was designed to combine the blockade of the PD-L1/PD-1 axis with conditional 4-1BB costimulation specifically confined to the tumor microenvironment (TME). The antibody demonstrated simultaneous binding to purified EGFR, PD-L1, and 4-1BB in solution, effective blockade of the PD-L1/PD1 interaction, and potent 4-1BB-mediated costimulation, but only in the presence of EGFR-expressing cells. These results demonstrate the feasibility of IgTT-4E1-S specifically blocking the PD-L1/PD-1 axis and inducing EGFR-conditional 4-1BB agonist activity.

12.
Theranostics ; 13(3): 1028-1041, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36793863

RESUMO

Despite the clinical success of the first bispecific antibody approved by the FDA against B cell malignancies (blinatumomab), many obstacles remain such as dosing, treatment resistance, and modest efficacy in solid tumors. To overcome these limitations, considerable efforts have been dedicated to the development of multispecific antibodies, opening up new avenues to address both the complex biology of cancer and the onset of anti-tumoral immune responses. Simultaneous targeting of two tumor-associated antigens is presumed to enhance cancer cell selectivity and reduce immune escape. Co-engagement of CD3, along with agonists of co-stimulatory molecules or antagonists of co-inhibitory immune checkpoint receptors in a single molecule, may revert T cell exhaustion. Similarly, targeting of two activating receptors in NK cells may improve their cytotoxic potency. And these are only examples of the potential of antibody-based molecular entities engaging three (or more) relevant targets. From the perspective of health care costs, multispecific antibodies are appealing, since a similar (or superior) therapeutic effect could be obtained with a single therapeutic agent as with a combination of different monoclonal antibodies. Despite challenges in production, multispecific antibodies are endowed with unprecedented properties, which may render them more potent biologics for cancer therapy.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Neoplasias , Humanos , Anticorpos Biespecíficos/farmacologia , Imunoterapia , Neoplasias/tratamento farmacológico , Células Matadoras Naturais , Antineoplásicos/farmacologia , Anticorpos Monoclonais/uso terapêutico
13.
Oncoimmunology ; 12(1): 2205336, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37114242

RESUMO

Immune checkpoint blockade (ICB) with antibodies has shown durable clinical responses in a wide range of cancer types, but the overall response rate is still limited. Other effective therapeutic modalities to increase the ICB response rates are urgently needed. New bispecific antibody (bsAb) formats combining the ICB effect and a direct action on cancer cells could improve the efficacy of current immunotherapies. Here, we report the development of a PD-L1/EGFR symmetric bsAb by fusing a dual-targeting tandem trimmer body with the human IgG1 hinge and Fc regions. The bsAb was characterized in vitro and the antitumor efficacy was evaluated in humanized mice bearing xenografts of aggressive triple-negative breast cancer and lung cancer. The IgG-like hexavalent bsAb, designated IgTT-1E, was able to simultaneously bind both EGFR and PD-L1 antigens, inhibit EGF-mediated proliferation, effectively block PD-1/PD-L1 interaction, and induce strong antigen-specific antibody-dependent cellular cytotoxicity activity in vitro. Potent therapeutic efficacies of IgTT-1E in two different humanized mouse models were observed, where tumor growth control was associated with a significantly increased proportion of CD8+ T cells. These results support the development of IgTT-1E for the treatment of EGFR+ cancers.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Humanos , Camundongos , Animais , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Antígeno B7-H1 , Linfócitos T CD8-Positivos , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Receptores ErbB
14.
Adv Sci (Weinh) ; 10(34): e2304818, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37863812

RESUMO

Administration of neutralizing antibodies (nAbs) has proved to be effective by providing immediate protection against SARS-CoV-2. However, dual strategies combining virus neutralization and immune response stimulation to enhance specific cytotoxic T cell responses, such as dendritic cell (DC) cross-priming, represent a promising field but have not yet been explored. Here, a broadly nAb, TNT , are first generated by grafting an anti-RBD biparatopic tandem nanobody onto a trimerbody scaffold. Cryo-EM data show that the TNT structure allows simultaneous binding to all six RBD epitopes, demonstrating a high-avidity neutralizing interaction. Then, by C-terminal fusion of an anti-DNGR-1 scFv to TNT , the bispecific trimerbody TNT DNGR-1 is generated to target neutralized virions to type 1 conventional DCs (cDC1s) and promote T cell cross-priming. Therapeutic administration of TNT DNGR-1, but not TNT , protects K18-hACE2 mice from a lethal SARS-CoV-2 infection, boosting virus-specific humoral responses and CD8+ T cell responses. These results further strengthen the central role of interactions with immune cells in the virus-neutralizing antibody activity and demonstrate the therapeutic potential of the Fc-free strategy that can be used advantageously to provide both immediate and long-term protection against SARS-CoV-2 and other viral infections.


Assuntos
Anticorpos Neutralizantes , COVID-19 , Camundongos , Animais , Anticorpos Neutralizantes/uso terapêutico , Linfócitos T Citotóxicos , SARS-CoV-2 , Apresentação Cruzada , Células Dendríticas
15.
Int Rev Cell Mol Biol ; 369: 71-87, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35777865

RESUMO

Trimerbodies, with their unique structural and functional properties, are the basis of a new generation of therapeutic antibodies, which due to their small size and plasticity are ideal for the generation of novel biological protein drugs with multiple competitive advantages over conventional full-length monoclonal antibodies. Since their emergence, trimerbodies have been used in preclinical cancer diagnosis and therapy. Trimerbodies are highly adaptable molecules, as they allow target-specific modulation of T cell-mediated anti-tumor immunity to enhance preexisting responses or to generate de novo immune responses. In fact, a tumor-specific humanized 4-1BB-agonistic trimerbody has shown a rather impressive safety and efficacy profile in preclinical studies making it a realistic option for clinical development. Moreover, thanks to the avidity effect they are endowed with considerable therapeutic potential as carriers to deliver cytotoxic payloads to tumors. In addition, molecular imaging studies could benefit from some intermediate-sized trivalent trimerbodies as promising candidates for targeted therapy and tumor imaging.


Assuntos
Anticorpos Monoclonais , Antineoplásicos Imunológicos , Imunoterapia , Neoplasias , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Humanos
16.
Oncoimmunology ; 11(1): 2034355, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154908

RESUMO

Retargeting of T lymphocytes toward cancer cells by bispecific antibodies has demonstrated its therapeutic potential, with one such antibody approved for the treatment of acute lymphoblastic leukemia (blinatumomab) and several other in clinical trials. However, improvement of their efficacy and selectivity for solid tumors is still required. Here, we describe a novel tandem T-cell recruiting trispecific antibody for the treatment of colorectal cancer (CRC). This construct, termed trispecific T-cell engager (TriTE), consists of a CD3-specific single-chain Fv (scFv) flanked by anti-epidermal growth factor receptor (EGFR) and anti-epithelial cell adhesion molecule (EpCAM) single-domain VHH antibodies. The TriTE was well expressed in mammalian and yeast cells, bound the cognate antigens of the three parental antibodies, and enabled the specific cytolysis of EGFR- and/or EpCAM-expressing cancer cells, without inducing T cell activation and cytoxicity against double-negative (EGFR-EpCAM-) cancer cells. Bivalent bispecific targeting of double-positive HCT116 cells by TriTE improved in vitro potency up to 100-fold compared to single-positive cells and significantly prolonged survival in vivo. In addition, it was less efficient at killing single-positive target cells than the corresponding bispecific controls, leading to potentially enhanced tumor specificity. Moreover, dual targeting of two tumor-associated antigens may contribute toward preventing the tumor escape by antigen loss caused by selective pressures from conventional single-targeting T-cell engagers, and may help to overcome antigenic heterogeneity.


Assuntos
Neoplasias Colorretais , Linfócitos T , Animais , Neoplasias Colorretais/tratamento farmacológico , Molécula de Adesão da Célula Epitelial , Receptores ErbB/metabolismo , Ativação Linfocitária , Mamíferos/metabolismo
17.
iScience ; 25(9): 104958, 2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36072551

RESUMO

Costimulation of tumor-infiltrating T lymphocytes by anti-4-1BB monoclonal antibodies (mAbs) has shown anti-tumor activity in human trials, but can be associated with significant off-tumor toxicities involving FcγR interactions. Here, we introduce albumin-fused mouse and human bispecific antibodies with clinically favorable pharmacokinetics designed to confine 4-1BB costimulation to the tumor microenvironment. These Fc-free 4-1BB agonists consist of an EGFR-specific VHH antibody, a 4-1BB-specific scFv, and a human albumin sequence engineered for high FcRn binding connected in tandem (LiTCo-Albu). We demonstrate in vitro cognate target engagement, EGFR-specific costimulatory activity, and FcRn-driven cellular recycling similar to non-fused FcRn high-binding albumin. The mouse LiTCo-Albu exhibited a prolonged circulatory half-life and in vivo tumor inhibition, with no indication of 4-1BB mAb-associated toxicity. Furthermore, we show a greater therapeutic effect when used in combination with PD-1-blocking mAbs. These findings demonstrate the feasibility of tumor-specific LiTCo-Albu antibodies for safe and effective costimulatory strategies in cancer immunotherapy.

18.
Cancer Immunol Res ; 10(4): 498-511, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35362043

RESUMO

Chimeric antigen receptor (CAR)-modified T cells have revolutionized the treatment of CD19-positive hematologic malignancies. Although anti-CD19 CAR-engineered autologous T cells can induce remission in patients with B-cell acute lymphoblastic leukemia, a large subset relapse, most of them with CD19-positive disease. Therefore, new therapeutic strategies are clearly needed. Here, we report a comprehensive study comparing engineered T cells either expressing a second-generation anti-CD19 CAR (CAR-T19) or secreting a CD19/CD3-targeting bispecific T-cell engager antibody (STAb-T19). We found that STAb-T19 cells are more effective than CAR-T19 cells at inducing cytotoxicity, avoiding leukemia escape in vitro, and preventing relapse in vivo. We observed that leukemia escape in vitro is associated with rapid and drastic CAR-induced internalization of CD19 that is coupled with lysosome-mediated degradation, leading to the emergence of transiently CD19-negative leukemic cells that evade the immune response of engineered CAR-T19 cells. In contrast, engineered STAb-T19 cells induce the formation of canonical immunologic synapses and prevent the CD19 downmodulation observed in anti-CD19 CAR-mediated interactions. Although both strategies show similar efficacy in short-term mouse models, there is a significant difference in a long-term patient-derived xenograft mouse model, where STAb-T19 cells efficiently eradicated leukemia cells, but leukemia relapsed after CAR-T19 therapy. Our findings suggest that the absence of CD19 downmodulation in the STAb-T19 strategy, coupled with the continued antibody secretion, allows an efficient recruitment of the endogenous T-cell pool, resulting in fast and effective elimination of cancer cells that may prevent CD19-positive relapses frequently associated with CAR-T19 therapies.


Assuntos
Leucemia , Linfócitos T , Animais , Antígenos CD19 , Humanos , Imunoterapia Adotiva/métodos , Camundongos , Recidiva
20.
Clin Cancer Res ; 27(11): 3167-3177, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33785484

RESUMO

PURPOSE: The induction of 4-1BB signaling by agonistic antibodies can drive the activation and proliferation of effector T cells and thereby enhance a T-cell-mediated antitumor response. Systemic administration of anti-4-1BB-agonistic IgGs, although effective preclinically, has not advanced in clinical development due to their severe hepatotoxicity. EXPERIMENTAL DESIGN: Here, we generated a humanized EGFR-specific 4-1BB-agonistic trimerbody, which replaces the IgG Fc region with a human collagen homotrimerization domain. It was characterized by structural analysis and in vitro functional studies. We also assessed pharmacokinetics, antitumor efficacy, and toxicity in vivo. RESULTS: In the presence of a T-cell receptor signal, the trimerbody provided potent T-cell costimulation that was strictly dependent on 4-1BB hyperclustering at the point of contact with a tumor antigen-displaying cell surface. It exhibits significant antitumor activity in vivo, without hepatotoxicity, in a wide range of human tumors including colorectal and breast cancer cell-derived xenografts, and non-small cell lung cancer patient-derived xenografts associated with increased tumor-infiltrating CD8+ T cells. The combination of the trimerbody with a PD-L1 blocker led to increased IFNγ secretion in vitro and resulted in tumor regression in humanized mice bearing aggressive triple-negative breast cancer. CONCLUSIONS: These results demonstrate the nontoxic broad antitumor activity of humanized Fc-free tumor-specific 4-1BB-agonistic trimerbodies and their synergy with checkpoint blockers, which may provide a way to elicit responses in most patients with cancer while avoiding Fc-mediated adverse reactions.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Receptores ErbB , Imunoterapia/métodos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/uso terapêutico , Animais , Neoplasias da Mama/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Neoplasias Pulmonares/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/fisiologia , Camundongos Transgênicos , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA