Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34172583

RESUMO

Costimulation via CD137 (4-1BB) enhances antitumor immunity mediated by cytotoxic T lymphocytes. Anti-CD137 agonist antibodies elicit mild liver inflammation in mice, and the maximum tolerated dose of Urelumab, an anti-human CD137 agonist monoclonal antibody, in the clinic was defined by liver inflammation-related side effects. A protease-activated prodrug form of the anti-mouse CD137 agonist antibody 1D8 (1D8 Probody therapeutic, Pb-Tx) was constructed and found to be selectively activated in the tumor microenvironment. This construct, which encompasses a protease-cleavable linker holding in place a peptide that masks the antigen binding site, exerted antitumor effects comparable to the unmodified antibody but did not result in liver inflammation. Moreover, it efficaciously synergized with both PD-1 blockade and adoptive T-cell therapy. Surprisingly, minimal active Pb-Tx reached tumor-draining lymph nodes, and regional lymphadenectomy did not abrogate antitumor efficacy. By contrast, S1P receptor-dependent recirculation of T cells was absolutely required for efficacy. The preferential cleavage of the anti-CD137 Pb-Tx by tumor proteases offers multiple therapeutic opportunities, including neoadjuvant therapy, as shown by experiments in which the Pb-Tx is given prior to surgery to avoid spontaneous metastases.


Assuntos
Antineoplásicos/toxicidade , Antineoplásicos/uso terapêutico , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos , Imunoterapia , Inflamação/patologia , Fígado/patologia , Neoplasias Pulmonares/secundário , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Camundongos , Terapia Neoadjuvante , Peptídeo Hidrolases/metabolismo
2.
Proc Natl Acad Sci U S A ; 112(24): 7551-6, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26034288

RESUMO

Cancer immunotherapy is undergoing significant progress due to recent clinical successes by refined adoptive T-cell transfer and immunostimulatory monoclonal Ab (mAbs). B16F10-derived OVA-expressing mouse melanomas resist curative immunotherapy with either adoptive transfer of activated anti-OVA OT1 CTLs or agonist anti-CD137 (4-1BB) mAb. However, when acting in synergistic combination, these treatments consistently achieve tumor eradication. Tumor-infiltrating lymphocytes that accomplish tumor rejection exhibit enhanced effector functions in both transferred OT-1 and endogenous cytotoxic T lymphocytes (CTLs). This is consistent with higher levels of expression of eomesodermin in transferred and endogenous CTLs and with intravital live-cell two-photon microscopy evidence for more efficacious CTL-mediated tumor cell killing. Anti-CD137 mAb treatment resulted in prolonged intratumor persistence of the OT1 CTL-effector cells and improved function with focused and confined interaction kinetics of OT-1 CTL with target cells and increased apoptosis induction lasting up to six days postadoptive transfer. The synergy of adoptive T-cell therapy and agonist anti-CD137 mAb thus results from in vivo enhancement and sustainment of effector functions.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Imunoterapia Adotiva/métodos , Melanoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Proteínas Aviárias/genética , Linhagem Celular Tumoral , Terapia Combinada , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/genética , Proteínas com Domínio T/metabolismo , Microambiente Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/deficiência , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
3.
Eur J Immunol ; 46(3): 513-22, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26773716

RESUMO

CD137 (4-1BB, TNF-receptor superfamily 9) is a surface glycoprotein of the TNFR family which can be induced on a variety of leukocyte subsets. On T and NK cells, CD137 is expressed following activation and, if ligated by its natural ligand (CD137L), conveys polyubiquitination-mediated signals via TNF receptor associated factor 2 that inhibit apoptosis, while enhancing proliferation and effector functions. CD137 thus behaves as a bona fide inducible costimulatory molecule. These functional properties of CD137 can be exploited in cancer immunotherapy by systemic administration of agonist monoclonal antibodies, which increase anticancer CTLs and enhance NK-cell-mediated antibody-dependent cell-mediated cytotoxicity. Reportedly, anti-CD137 mAb and adoptive T-cell therapy strongly synergize, since (i) CD137 expression can be used to select the T cells endowed with the best activities against the tumor, (ii) costimulation of the lymphocyte cultures to be used in adoptive T-cell therapy can be done with CD137 agonist antibodies or CD137L, and (iii) synergistic effects upon coadministration of T cells and antibodies are readily observed in mouse models. Furthermore, the signaling cytoplasmic tail of CD137 is a key component of anti-CD19 chimeric antigen receptors that are used to redirect T cells against leukemia and lymphoma in the clinic. Ongoing phase II clinical trials with agonist antibodies and the presence of CD137 sequence in these successful chimeric antigen receptors highlight the importance of CD137 in oncoimmunology.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia Adotiva , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Citotóxicos/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 , Animais , Citotoxicidade Celular Dependente de Anticorpos , Ensaios Clínicos Fase II como Assunto , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Camundongos , Transdução de Sinais
4.
Cancer Immunol Immunother ; 65(5): 493-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26970765

RESUMO

CD137(4-1BB) costimulation and adoptive T cell therapy strongly synergize in terms of achieving maximal efficacy against experimental cancers. These costimulatory biological functions of CD137 have been exploited by means of introducing the CD137 signaling domain in clinically successful chimeric antigen receptors and to more efficiently expand T cells in culture. In addition, immunomagnetic sorting of CD137-positive T cells among tumor-infiltrating lymphocytes selects for the fittest antitumor T lymphocytes for subsequent cultures. In mouse models, co-infusion of both agonist antibodies and T cells attains marked synergistic effects that result from more focused and intense cytolytic activity visualized under in vivo microscopy and from more efficient entrance of T cells into the tumor through the vasculature. These several levels of dynamic interaction between adoptive T cell therapy and CD137 offer much opportunity to raise the efficacy of current cancer immunotherapies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/transplante , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Terapia Combinada , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/transplante , Modelos Imunológicos , Neoplasias/imunologia , Neoplasias/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/transplante , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
5.
Clin Cancer Res ; 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38630754

RESUMO

PURPOSE: Cancer patients frequently undergo radiotherapy in their clinical management with unintended irradiation of blood vessels and copiously irrigated organs in which polymorphonuclear leukocytes circulate. Following the observation that such low doses of ionizing radiation are able to induce neutrophils to extrude neutrophil extracellular traps (NETs), we have investigated the mechanisms, consequences and the occurrence of such phenomena in patients undergoing radiotherapy. EXPERIMENTAL DESIGN: NETosis was analyzed in cultures of neutrophils isolated from healthy donors, cancer patients and cancer-bearing mice under confocal microscopy. Cocultures of radiation-induced NETs, immune effector lymphocytes and tumor cells were used to study the effects of irradiation-induced NETs on immune cytotoxicity. Radiation-induced NETs were intravenously injected to mice assessing their effects on metastasis. Circulating NETs in irradiated cancer patients were measured by ELISA methods detecting MPO-DNA complexes and citrullinated H3. RESULTS: Very low γ-radiation doses (0.5-1 Gy) given to neutrophils elicit NET formation in a manner dependent on oxidative stress, NADPH oxidase activity and autocrine interleukin-8. Radiation-induced NETs interfere with NK- and T-cell cytotoxicity. As a consequence, pre-injection of irradiation-induced NETs increases the number of successful metastases in mouse tumor models. Increases in circulating NETs were readily detected in two prospective series of patients following the first fraction of their radiotherapy courses. CONCLUSIONS: NETosis is induced by low-dose ionizing irradiation in a neutrophil-intrinsic fashion and radiation-induced NETs are able to interfere with immune-mediated cytotoxicity. Radiation-induced NETs foster metastasis in mouse models and can be detected in the circulation of patients undergoing conventional radiotherapy treatments.

6.
J Immunol ; 187(11): 6130-42, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22048768

RESUMO

Twenty-four patients with metastatic cancer received two cycles of four daily immunizations with monocyte-derived dendritic cells (DC). DC were incubated with preheated autologous tumor lysate and subsequently with IFN-α, TNF-α, and polyinosinic:polycytidylic acid to attain type 1 maturation. One DC dose was delivered intranodally, under ultrasound control, and the rest intradermally in the opposite thigh. Cyclophosphamide (day -7), GM-CSF (days 1-4), and pegIFN alpha-2a (days 1 and 8) completed each treatment cycle. Pretreatment with cyclophosphamide decreased regulatory T cells to levels observed in healthy subjects both in terms of percentage and in absolute counts in peripheral blood. Treatment induced sustained elevations of IL-12 in serum that correlated with the output of IL-12p70 from cultured DC from each individual. NK activity in peripheral blood was increased and also correlated with the serum concentration of IL-12p70 in each patient. Circulating endothelial cells decreased in 17 of 18 patients, and circulating tumor cells markedly dropped in 6 of 19 cases. IFN-γ-ELISPOT responses to DC plus tumor lysate were observed in 4 of 11 evaluated cases. Tracing DC migration with [(111)In] scintigraphy showed that intranodal injections reached deeper lymphatic chains in 61% of patients, whereas with intradermal injections a small fraction of injected DC was almost constantly shown to reach draining inguinal lymph nodes. Five patients experienced disease stabilization, but no objective responses were documented. This combinatorial immunotherapy strategy is safe and feasible, and its immunobiological effects suggest potential activity in patients with minimal residual disease. A randomized trial exploring this hypothesis is currently ongoing.


Assuntos
Antígenos de Neoplasias/imunologia , Antineoplásicos/uso terapêutico , Células Dendríticas/transplante , Imunoterapia/métodos , Neoplasias/terapia , Adjuvantes Imunológicos/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Separação Celular , Ciclofosfamida/uso terapêutico , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Interferon-alfa/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Projetos Piloto , Polietilenoglicóis/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
7.
Expert Opin Ther Targets ; 27(11): 1035-1042, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37993172

RESUMO

INTRODUCTION: Interleukin-18 (IL-18) is a myeloid leukocyte inflammatory mediator whose main known function is to elicit IFNγ secretion from T and NK cells. AREAS COVERED: This function offers potential in cancer immunotherapy but as a single treatment, preclinical and clinical antitumor activities are modest. IL-18 bioactivity is chiefly downregulated by a decoy soluble receptor named IL18-binding protein (IL-18BP) that is induced by IFNγ as a negative feedback mechanism. Recent advances indicate promising efficacy of IL-18 at armoring CAR-T cells for the treatment of hematological malignancies. Preclinical research has also yielded IL-18 constructs that do not bind IL-18BP but have preserved activity on the receptor and exert markedly increased antitumor effects. Indeed, agents of this kind are undergoing clinical trials. The synergistic effects of IL-18 and IL-12 in combination to induce IFNγ are extremely potent but are toxic if systemically delivered. In mouse models, IL-12 and decoy-resistant variants of IL-18 can be efficaciously used as local treatments for tumors by exploiting mRNA intratumoral co-delivery. Moreover, antitumor T cells can be transiently engineered with mRNAs encoding this combination of cytokines to attain efficacious synergistic effects also upon intratumoral delivery. EXPERT OPINION: IL-18 certainly holds promise for immunotherapy in combination with other agents and for local approaches.


Assuntos
Interleucina-18 , Neoplasias , Animais , Camundongos , Imunoterapia , Neoplasias/terapia , Interleucina-12 , Citocinas
8.
Mol Ther Nucleic Acids ; 33: 668-682, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37650116

RESUMO

Intratumoral immunotherapy strategies for cancer based on interleukin-12 (IL-12)-encoding cDNA and mRNA are under clinical development in combination with anti-PD-(L)1 monoclonal antibodies. To make the most of these approaches, we have constructed chimeric mRNAs encoding single-chain IL-12 fused to single-chain fragment variable (scFv) antibodies that bind to transforming growth factor ß (TGF-ß) and CD137 (4-1BB). Several neutralizing TGF-ß agents and CD137 agonists are also undergoing early-phase clinical trials. To attain TGF-ß and CD137 binding by the constructions, we used bispecific tandem scFv antibodies (taFvs) derived from the specific 1D11 and 1D8 monoclonal antibodies (mAbs), respectively. Transfection of mRNAs encoding the chimeric constructs achieved functional expression of the proteins able to act on their targets. Upon mRNA intratumoral injections in the transplantable mouse cancer models CT26, MC38, and B16OVA, potent therapeutic effects were observed following repeated injections into the tumors. Efficacy was dependent on the number of CD8+ T cells able to recognize tumor antigens that infiltrated the malignant tissue. Although the abscopal effects on concomitant uninjected lesions were modest, such distant effects on untreated lesions were markedly increased when combined with systemic PD-1 blockade.

9.
Cell Rep Med ; : 100978, 2023 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-36933554

RESUMO

Interleukin-12 (IL-12) gene transfer enhances the therapeutic potency of adoptive T cell therapies. We previously reported that transient engineering of tumor-specific CD8 T cells with IL-12 mRNA enhanced their systemic therapeutic efficacy when delivered intratumorally. Here, we mix T cells engineered with mRNAs to express either single-chain IL-12 (scIL-12) or an IL-18 decoy-resistant variant (DRIL18) that is not functionally hampered by IL-18 binding protein (IL-18BP). These mRNA-engineered T cell mixtures are repeatedly injected into mouse tumors. Pmel-1 T cell receptor (TCR)-transgenic T cells electroporated with scIL-12 or DRIL18 mRNAs exert powerful therapeutic effects in local and distant melanoma lesions. These effects are associated with T cell metabolic fitness, enhanced miR-155 control on immunosuppressive target genes, enhanced expression of various cytokines, and changes in the glycosylation profile of surface proteins, enabling adhesiveness to E-selectin. Efficacy of this intratumoral immunotherapeutic strategy is recapitulated in cultures of tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T cells on IL-12 and DRIL18 mRNA electroporation.

10.
Oncoimmunology ; 12(1): 2197370, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37035637

RESUMO

BO-112 is a poly I:C-based viral mimetic that exerts anti-tumor efficacy when intratumorally delivered in mouse models. Intratumoral BO-112 synergizes in mice with systemic anti-PD-1 mAbs and this combination has attained efficacy in PD1-refractory melanoma patients. We sought to evaluate the anti-tumor efficacy of BO-112 pre-surgically applied in neoadjuvant settings to mouse models. We have observed that repeated intratumoral injections of BO-112 prior to surgical excision of the primary tumor significantly reduced tumor metastasis from orthotopically implanted 4T1-derived tumors and subcutaneous MC38-derived tumors in mice. Such effects were enhanced when combined with systemic anti-PD-1 mAb. The anti-tumor efficacy of this neoadjuvant immunotherapy approach depended on the presence of antigen-specific effector CD8 T cells and cDC1 antigen-presenting cells. Since BO-112 has been successful in phase-two clinical trials for metastatic melanoma, these results provide a strong rationale for translating this pre-surgical strategy into clinical settings, especially in combination with standard-of-care checkpoint inhibitors.


Assuntos
Melanoma , Terapia Neoadjuvante , Animais , Camundongos , Linfócitos T , Imunoterapia/métodos , Melanoma/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Adjuvantes Imunológicos
11.
Cancer Immunol Res ; 11(2): 184-198, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36478221

RESUMO

IL12-based local gene therapy of cancer constitutes an active area of clinical research using plasmids, mRNAs, and viral vectors. To improve antitumor effects, we have experimentally tested the combination of mRNA constructs encoding IL12 and IL18. Moreover, we have used a form of IL18 [decoy-resistant IL18 (DR-18)] which has preserved bioactivity but does not bind to the IL18 binding protein decoy receptor. Both cytokines dramatically synergize to induce IFNγ release from mouse splenocytes, and, if systemically cotransferred to the liver, they mediate lethal toxicity. However, if given intratumorally to B16OVA tumor-bearing mice, the combination attains efficacy against the directly treated tumor and moderate tumor-delaying activity on distant noninjected lesions. Cotreatment was conducive to the presence of more activated CD8+ T cells in the treated and noninjected tumors. In keeping with these findings, the efficacy of treatment was contingent on the integrity of CD8+ T cells and cDC1 dendritic cells in the treated mice. Furthermore, efficacy of IL12 plus DR-18 local mRNA coinjection against distant concomitant tumors could be enhanced upon combination with anti-PD-1 mAb systemic treatment, thus defining a feasible synergistic immunotherapy strategy.


Assuntos
Interleucina-18 , Neoplasias , Animais , Camundongos , Neoplasias/genética , Neoplasias/terapia , Linfócitos T CD8-Positivos , Imunoterapia , Interleucina-12/metabolismo
12.
Mol Ther Nucleic Acids ; 33: 599-616, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37637207

RESUMO

IL-12 is a potent cytokine for cancer immunotherapy. However, its systemic delivery as a recombinant protein has shown unacceptable toxicity in the clinic. Currently, the intratumoral injection of IL-12-encoding mRNA or DNA to avoid such side effects is being evaluated in clinical trials. In this study, we aimed to improve this strategy by further favoring IL-12 tethering to the tumor. We generated in vitro transcribed mRNAs encoding murine single-chain IL-12 fused to diabodies binding to CSF1R and/or PD-L1. These targeted molecules are expressed in the tumor microenvironment, especially on myeloid cells. The binding capacity of chimeric constructs and the bioactivity of IL-12 were demonstrated in vitro and in vivo. Doses as low as 0.5 µg IL-12-encoding mRNA achieved potent antitumor effects in subcutaneously injected B16-OVA and MC38 tumors. Treatment delivery was associated with increases in IL-12p70 and IFN-γ levels in circulation. Fusion of IL-12 to the diabodies exerted comparable efficacy against bilateral tumor models. However, it achieved tethering to myeloid cells infiltrating the tumor, resulting in nearly undetectable systemic levels of IL-12 and IFN-γ. Overall, tethering IL-12 to intratumoral myeloid cells in the mRNA-transferred tumors achieves similar efficacy while reducing the dangerous systemic bioavailability of IL-12.

13.
Expert Opin Drug Discov ; 17(1): 41-53, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34496689

RESUMO

INTRODUCTION: Immune checkpoint inhibitors and adoptive T-cell therapy based on chimeric antigen receptors are the spearhead strategies to exploit the immune system to fight cancer. To take advantage of the full potential of the immune system, cancer immunotherapy must incorporate new biotechnologies such as mRNA technology that may synergize with already approved immunotherapies and act more effectively on immune targets. AREAS COVERED: This review describes the basics of mRNA biotechnology and provides insight into the recent advances in the use of mRNA for the local and systemic delivery of immunostimulatory antibodies, proinflammatory cytokines or for optimizing adoptive T-cell therapy. EXPERT OPINION: mRNA-based nanomedicines have great potential to expand the arsenal of immunotherapy tools due to their ability to simplify and accelerate drug development and their suitability for transient and local expression of immunostimulatory molecules, whose systemic and sustained expression would be toxic. The success of mRNA-based COVID-19 vaccines has highlighted the feasibility of this approach. Continuous advances in the delivery and construction of RNA-based vectors hold promise for improvements in clinical efficacy.


Assuntos
COVID-19 , Neoplasias , Vacinas contra COVID-19 , Descoberta de Drogas , Humanos , Imunoterapia , Neoplasias/tratamento farmacológico , Neoplasias/genética , RNA Mensageiro/genética , SARS-CoV-2
14.
Pharmacol Ther ; 239: 108189, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35430292

RESUMO

Interleukin-12 is considered a potent agent to enhance antitumor immune responses. It belongs to a family of heterodimeric cytokines with key roles in the up-regulation and down-regulation of cellular immunity. Since its discovery, recombinant IL-12 was found to exert potent antitumor effects in rodent tumor models and was rapidly tested in the clinic with an unfavorable benefit/toxicity profile. Localized delivery of IL-12 dramatically improves the therapeutic index and this approach is being applied in the clinic based on in-vivo electroporation of naked plasmid DNA encoding IL-12, mRNA formulations, viral vectors and tumor-targeted fusion proteins. Other biotechnology strategies such as IL-12-engineered local adoptive cell therapy and pro-cytokines can also be used to improve results and broaden the therapeutic window. Combination strategies of such localized IL-12-based approaches with checkpoint inhibitors are yielding promising results both preclinically and in the early-phase clinical trials.


Assuntos
Interleucina-12 , Neoplasias , Humanos , Interleucina-12/genética , Imunoterapia/métodos , Vetores Genéticos , Imunoterapia Adotiva , Fatores Imunológicos , Neoplasias/terapia
15.
Cancer Discov ; 12(9): 2140-2157, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-35771565

RESUMO

Interleukin-8 (CXCL8) produced in the tumor microenvironment correlates with poor response to checkpoint inhibitors and is known to chemoattract and activate immunosuppressive myeloid leukocytes. In human cancer, IL8 mRNA levels correlate with IL1B and TNF transcripts. Both cytokines induced IL-8 functional expression from a broad variety of human cancer cell lines, primary colon carcinoma organoids, and fresh human tumor explants. Although IL8 is absent from the mouse genome, a similar murine axis in which TNFα and IL-1ß upregulate CXCL1 and CXCL2 in tumor cells was revealed. Furthermore, intratumoral injection of TNFα and IL-1ß induced IL-8 release from human malignant cells xenografted in immunodeficient mice. In all these cases, the clinically used TNFα blockers infliximab and etanercept or the IL-1ß inhibitor anakinra was able to interfere with this pathogenic cytokine loop. Finally, in paired plasma samples of patients with cancer undergoing TNFα blockade with infliximab in a clinical trial, reductions of circulating IL-8 were substantiated. SIGNIFICANCE: IL-8 attracts immunosuppressive protumor myeloid cells to the tumor microenvironment, and IL-8 levels correlate with poor response to checkpoint inhibitors. TNFα and IL-1ß are identified as major inducers of IL-8 expression on malignant cells across cancer types and models in a manner that is druggable with clinically available neutralizing agents. This article is highlighted in the In This Issue feature, p. 2007.


Assuntos
Citocinas , Fator de Necrose Tumoral alfa , Animais , Citocinas/metabolismo , Humanos , Infliximab/farmacologia , Infliximab/uso terapêutico , Interleucina-1beta/metabolismo , Interleucina-8/genética , Camundongos , Microambiente Tumoral , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
16.
Int J Cancer ; 129(2): 374-86, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20853321

RESUMO

Anti-CTLA-4 monoclonal antibodies (mAb) that block the interaction of CTLA-4 with CD80 and CD86 such as tremelimumab and ipilimumab are currently being tested in the clinic for cancer treatment exploiting their properties to de-repress tumor-specific cellular immunity. Addition of the fully human anti-CTLA-4 (tremelimumab) to cultures of human T cells with allogenic dendritic cells (DCs) did not increase proliferation. Magnetic bead-mediated elimination of CD4(+) CD25(+) regulatory T cells (T(reg)) before setting up those alloreactive cultures also largely failed to increase primary proliferation. In contrast, predepletion of CD4(+) CD25(+) T(reg) and culture in the presence of tremelimumab synergistically resulted in increased proliferation and DC:T-cell aggregation. These effects were much more prominent in CD4 than in CD8 T cells. The synergy mechanism can be traced to enhanced CTLA-4 expression in effector cells as a result of T(reg) elimination, thereby offering more targets to the blocking antibody. Human T cells and allogenic DCs (derived both from healthy donors and advanced cancer patients) were coinjected in the peritoneum of Rag2(-/-) IL-2Rγ(-/-) mice. In these conditions, tremelimumab injected intravenously did not significantly enhance alloreactive proliferation unless T(reg) cells had been predepleted. Synergistic effects in vivo were again largely restricted to the CD4 T-cell compartment. In addition, T(reg) depletion and CTLA-4 blockade synergistically enhanced specific cytotoxicity raised in culture against autologous EBV-transformed cell lines. Taken together, these experiments indicate that tremelimumab therapy may benefit from previous or concomitant T(reg) depletion.


Assuntos
Anticorpos Bloqueadores , Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Fatores Imunológicos/farmacologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais Humanizados , Antígeno CTLA-4 , Proliferação de Células/efeitos dos fármacos , Transplante de Células , Células Cultivadas , Células Dendríticas/transplante , Humanos , Camundongos , Camundongos Transgênicos , Linfócitos T/imunologia , Linfócitos T/transplante , Linfócitos T Reguladores/efeitos dos fármacos
17.
Liver Int ; 31(10): 1542-53, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22093330

RESUMO

BACKGROUND: Diverse oncolytic viruses (OV) are being designed for the treatment of cancer. The characteristics of the parental virus strains may influence the properties of these agents. AIMS: To characterize two herpes simplex virus 1 strains (HSV-1 17syn(+) and HFEM) as platforms for virotherapy against liver cancer. METHODS: The luciferase reporter gene was introduced in the intergenic region 20 locus of both HSV-1 strains, giving rise to the Cgal-Luc and H6-Luc viruses. Their properties were studied in hepatocellular carcinoma (HCC) cells in vitro. Biodistribution was monitored by bioluminescence imaging (BLI) in athymic mice and immune-competent Balb/c mice. Immunogenicity was studied by MHC-tetramer staining, in vivo killing assays and determination of specific antibody production. Intratumoural transgene expression and oncolytic effect were studied in HuH-7 xenografts. RESULTS: The H6-Luc virus displayed a syncytial phenotype and showed higher cytolytic effect on some HCC cells. Upon intravenous or intrahepatic injection in mice, both viruses showed a transient transduction of the liver with rapid relocalization of bioluminescence in adrenal glands, spinal cord, uterus and ovaries. No significant differences were observed in the immunogenicity of these viruses. Local intratumoural administration caused progressive increase in transgene expression during the first 5 days and persisted for at least 2 weeks. H6-Luc achieved faster amplification of transgene expression and stronger inhibition of tumour growth than Cgal-Luc, although toxicity of these non-attenuated viruses should be reduced to obtain a therapeutic effect. CONCLUSIONS: The syncytial H6-Luc virus has a strong oncolytic potential on human HCC xenografts and could be the basis for potent OV.


Assuntos
Carcinoma Hepatocelular/terapia , Herpesvirus Humano 1/genética , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica/métodos , Animais , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Sobrevivência Celular , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Herpesvirus Humano 1/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Neoplasias Hepáticas/virologia , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Reação em Cadeia da Polimerase , Estatísticas não Paramétricas , Transdução Genética , Transgenes/genética
18.
Nat Commun ; 12(1): 7296, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911975

RESUMO

CD137 (4-1BB; TNFSR9) is an activation-induced surface receptor that through costimulation effects provide antigen-primed T cells with augmented survival, proliferation and effector functions as well as metabolic advantages. These immunobiological mechanisms are being utilised for cancer immunotherapy with agonist CD137-binding and crosslinking-inducing agents that elicit CD137 intracellular signaling. In this study, side-by-side comparisons show that provision of CD137 costimulation in-cis with regard to the TCR-CD3-ligating cell is superior to that provided in-trans in terms of T cell activation, proliferation, survival, cytokine secretion and mitochondrial fitness in mouse and human. Cis ligation of CD137 relative to the TCR-CD3 complex results in more intense canonical and non-canonical NF-κB signaling and provides a more robust induction of cell cycle and DNA damage repair gene expression programs. Here we report that the superiority of cis versus trans CD137-costimulation is readily observed in vivo and is relevant for understanding the immunotherapeutic effects of CAR T cells and CD137 agonistic therapies currently undergoing clinical trials, which may provide costimulation either in cis or in trans.


Assuntos
Complexo CD3/imunologia , Linfócitos T CD8-Positivos/imunologia , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Complexo CD3/genética , Proliferação de Células , Citocinas/genética , Citocinas/imunologia , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
19.
Cell Mol Immunol ; 17(6): 576-586, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32433539

RESUMO

Gene engineering and combinatorial approaches with other cancer immunotherapy agents may confer capabilities enabling full tumor rejection by adoptive T cell therapy (ACT). The provision of proper costimulatory receptor activity and cytokine stimuli, along with the repression of inhibitory mechanisms, will conceivably make the most of these treatment strategies. In this sense, T cells can be genetically manipulated to become refractory to suppressive mechanisms and exhaustion, last longer and differentiate into memory T cells while endowed with the ability to traffic to malignant tissues. Their antitumor effects can be dramatically augmented with permanent or transient gene transfer maneuvers to express or delete/repress genes. A combination of such interventions seeks the creation of the ultimate bionic T cell, perfected to seek and destroy cancer cells upon systemic or local intratumor delivery.


Assuntos
Biônica , Engenharia Genética , Transdução de Sinais , Linfócitos T/imunologia , Animais , Humanos , Imunidade/imunologia , Neoplasias/imunologia
20.
Clin Cancer Res ; 26(6): 1203-1204, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32001482

RESUMO

T-cell recognizable p53 hotspot mutations offer opportunities for immunotherapy and immune monitoring. Recognition of p53 mutations by peripheral blood CD8 and CD4 T lymphocytes has been revealed.See related article by Malekzadeh et al., p. 1267.


Assuntos
Antígenos de Neoplasias , Proteína Supressora de Tumor p53 , Antígenos de Neoplasias/genética , Linfócitos T CD4-Positivos/metabolismo , Humanos , Imunoterapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA