Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Ther ; 27(10): 1825-1835, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31331813

RESUMO

Chimeric antigen receptor (CAR)-engineered T cells are efficacious in controlling advanced leukemia and lymphoma, however, they fail in the treatment of solid cancer, which is thought to be due to insufficient T cell activation. We revealed that the immune response of CAR T cells with specificity for carcinoembryonic antigen (CEA) was more efficacious against CEA+ cancer cells when simultaneously incubated with an anti-CD30 immunotoxin or anti-CD30 CAR T cells, although the targeted cancer cells lack CD30. The same effect was achieved when the anti-CD30 single-chain variable fragment (scFv) was integrated into the extracellular domain of the anti-CEA CAR. Improvement in T cell activation was due to interfering with the T cell CD30-CD30L interaction by the antagonistic anti-CD30 scFv HRS3; an agonistic anti-CD30 scFv or targeting the high-affinity interleukin-2 (IL-2) receptor was not effective. T cells with the anti-CD30/CEA CAR showed superior immunity against established CEA+ CD30- tumors in a mouse model. The concept is broadly applicable since anti-CD30/TAG72 CAR T cells also showed improved elimination of TAG72+ CD30- cancer cells. Taken together, targeting CD30 on CAR T cells by the HRS3 scFv within the anti-tumor CAR improves the redirected immune response against solid tumors.


Assuntos
Antígeno Carcinoembrionário/imunologia , Neoplasias do Colo/terapia , Imunoterapia Adotiva/métodos , Antígeno Ki-1/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Células HEK293 , Humanos , Camundongos , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Ther ; 26(9): 2218-2230, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30055872

RESUMO

Adoptive cell therapy with chimeric antigen receptor (CAR)-redirected T cells induced spectacular regressions of leukemia and lymphoma, however, failed so far in the treatment of solid tumors. A cause is thought to be T cell repression through TGF-ß, which is massively accumulating in the tumor tissue. Here, we show that T cells with a CD28-ζ CAR, but not with a 4-1BB-ζ CAR, resist TGF-ß-mediated repression. Mechanistically, LCK activation and consequently IL-2 release and autocrine IL-2 receptor signaling mediated TGF-ß resistance; deleting the LCK-binding motif in the CD28 CAR abolished both IL-2 secretion and TGF-ß resistance, while IL-2 add-back restored TGF-ß resistance. Other γ-cytokines like IL-7 and IL-15 could replace IL-2 in this context. This is demonstrated by engineering IL-2 deficient CD28ΔLCK-ζ CAR T cells with a hybrid IL-7 receptor to provide IL-2R ß chain signaling upon IL-7 binding. Such modified T cells showed improved CAR T cell activity against TGF-ß+ tumors. Data draw the concept that an autocrine loop resulting in IL-2R signaling can make CAR T cells more potent in staying active against TGF-ß+ solid tumors.


Assuntos
Antígenos CD28/metabolismo , Interleucina-2/metabolismo , Interleucina-7/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos CD28/genética , Linhagem Celular , Células Cultivadas , Granzimas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Receptores de Antígenos Quiméricos/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
3.
Blood ; 128(13): 1711-22, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27535994

RESUMO

Adoptive cell therapy of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR)-modified T cells targeting CD19 induced lasting remission of this refractory disease in a number of patients. However, the treatment is associated with prolonged "on-target off-tumor" toxicities due to the targeted elimination of healthy B cells demanding more selectivity in targeting CLL cells. We identified the immunoglobulin M Fc receptor (FcµR), also known as the Fas apoptotic inhibitory molecule-3 or TOSO, as a target for a more selective treatment of CLL by CAR T cells. FcµR is highly and consistently expressed by CLL cells; only minor levels are detected on healthy B cells or other hematopoietic cells. T cells with a CAR specific for FcµR efficiently responded toward CLL cells, released a panel of proinflammatory cytokines and lytic factors, like soluble FasL and granzyme B, and eliminated the leukemic cells. In contrast to CD19 CAR T cells, anti-FcµR CAR T cells did not attack healthy B cells. T cells with anti-FcµR CAR delayed outgrowth of Mec-1-induced leukemia in a xenograft mouse model. T cells from CLL patients in various stages of the disease, modified by the anti-FcµR CAR, purged their autologous CLL cells in vitro without reducing the number of healthy B cells, which is the case with anti-CD19 CAR T cells. Compared with the currently used therapies, the data strongly imply a superior therapeutic index of anti-FcµR CAR T cells for the treatment of CLL.


Assuntos
Imunoterapia Adotiva/métodos , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Fc/antagonistas & inibidores , Receptores Fc/imunologia , Linfócitos T/imunologia , Adulto , Idoso , Animais , Linfócitos B/imunologia , Engenharia Celular , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Engenharia de Proteínas , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Immunol Rev ; 257(1): 83-90, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24329791

RESUMO

Adoptive T-cell therapy recently achieved impressive efficacy in early phase trials, in particular in hematologic malignancies, strongly supporting the notion that the immune system can control cancer. A current strategy of favor is based on ex vivo-engineered patient T cells, which are redirected by a chimeric antigen receptor (CAR) and recognize a predefined target by an antibody-derived binding domain. Such CAR T cells can substantially reduce the tumor burden as long as the targeted antigen is present on the cancer cells. However, given the tremendous phenotypic diversity in solid tumor lesions, a reasonable number of cancer cells are not recognized by a given CAR, considerably reducing the therapeutic success. This article reviews a recently described strategy for overcoming this shortcoming of the CAR T-cell therapy by modulating the tumor stroma by a CAR T-cell-secreted transgenic cytokine like interleukin-12 (IL-12). The basic process is that CAR T cells, when activated by their CAR, deposit IL-12 in the targeted tumor lesion, which in turn attracts an innate immune cell response toward those cancer cells that are invisible to CAR T cells. Such TRUCKs, T cells redirected for universal cytokine-mediated killing, exhibited remarkable efficacy against solid tumors with diverse cancer cell phenotypes, suggesting their evaluation in clinical trials.


Assuntos
Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Citocinas/metabolismo , Citotoxicidade Imunológica , Humanos , Imunidade Inata , Fatores Imunológicos/genética , Fatores Imunológicos/metabolismo , Imunomodulação , Imunoterapia Adotiva , Interleucina-12/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Células Estromais/metabolismo
5.
Mol Ther ; 24(8): 1423-34, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27112062

RESUMO

Recent clinical trials with chimeric antigen receptor (CAR) redirected T cells targeting CD19 revealed particular efficacy in the treatment of leukemia/lymphoma, however, were accompanied by a lasting depletion of healthy B cells. We here explored CD30 as an alternative target, which is validated in lymphoma therapy and expressed by a broad variety of Hodgkin's and non-Hodgkin's lymphomas. As a safty concern, however, CD30 is also expressed by lymphocytes and hematopoietic stem and progenitor cells (HSPCs) during activation. We revealed that HRS3scFv-derived CAR T cells are superior since they were not blocked by soluble CD30 and did not attack CD30(+) HSPCs while eliminating CD30(+) lymphoma cells. Consequently, normal hemato- and lymphopoiesis was not affected in the long-term in the humanized mouse; the number of blood B and T cells remained unchanged. We provide evidence that the CD30(+) HSPCs are protected against a CAR T-cell attack by substantially lower CD30 levels than lymphoma cells and higher levels of the granzyme B inactivating SP6/PI9 serine protease, which furthermore increased upon activation. Taken together, adoptive cell therapy with anti-CD30 CAR T cells displays a superior therapeutic index in the treatment of CD30(+) malignancies leaving healthy activated lymphocytes and HSPCs unaffected.

6.
Am J Pathol ; 182(6): 2121-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23562272

RESUMO

Cellular immunotherapy may provide a strategy to overcome the poor prognosis of metastatic and recurrent rhabdomyosarcoma (RMS) under the current regimen of polychemotherapy. Because little is known about resistance mechanisms of RMS to cytotoxic T cells, we investigated RMS cell lines and biopsy specimens for expression and function of immune costimulatory receptors and anti-apoptotic molecules by RT-PCR, Western blot analysis, IHC, and cytotoxicity assays using siRNA or transfection-modified RMS cell lines, together with engineered RMS-directed cytotoxic T cells specific for the fetal acetylcholine receptor. We found that costimulatory CD80 and CD86 were consistently absent from all RMSs tested, whereas inducible T-cell co-stimulator ligand (ICOS-L; alias B7H2) was expressed by a subset of RMSs and was inducible by tumor necrosis factor α in two of five RMS cell lines. Anti-apoptotic survivin, along with other inhibitor of apoptosis (IAP) family members (cIAP1, cIAP2, and X-linked inhibitor of apoptosis protein), was overexpressed by RMS cell lines and biopsy specimens. Down-regulation of survivin by siRNA or pharmacologically in RMS cells increased their susceptibility toward a T-cell attack, whereas induction of ICOS-L did not. Treatment of RMS-bearing Rag(-/-) mice with fetal acetylcholine receptor-specific chimeric T cells delayed xenograft growth; however, this happened without definitive tumor eradication. Combined blockade of survivin and application of chimeric T cells in vivo suppressed tumor proliferation during survivin inhibition. In conclusion, survivin blockade provides a strategy to sensitize RMS cells for T-cell-based therapy.


Assuntos
Proteínas Inibidoras de Apoptose/imunologia , Receptores Colinérgicos/imunologia , Rabdomiossarcoma/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Biópsia , Antígenos CD28/imunologia , Pré-Escolar , Receptores Coestimuladores e Inibidores de Linfócitos T/metabolismo , Citotoxicidade Imunológica/imunologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imunoterapia Adotiva/métodos , Ligante Coestimulador de Linfócitos T Induzíveis/biossíntese , Ligante Coestimulador de Linfócitos T Induzíveis/imunologia , Lactente , Proteínas Inibidoras de Apoptose/genética , Ligantes , Masculino , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Rabdomiossarcoma/patologia , Rabdomiossarcoma/prevenção & controle , Transdução de Sinais/imunologia , Survivina , Linfócitos T/transplante , Transplante Heterólogo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/imunologia
7.
Mol Ther ; 21(12): 2268-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23985696

RESUMO

Cytokine-induced killer (CIK) cells raised interest for use in cellular antitumor therapy due to their capability to recognize and destroy autologous tumor cells in a HLA-independent fashion. The antitumor attack of CIK cells, predominantly consisting of terminally differentiated CD8(+)CD56(+) cells, can be improved by redirecting by a chimeric antigen receptor (CAR) that recognizes the tumor cell and triggers CIK cell activation. The requirements for CIK cell activation were, however, so far less explored and are likely to be different from those of "younger" T cells. We revealed that CD28 and OX40 CARs produced higher interferon- secretion as compared with the first-generation ζ-CAR; CD28-ζ and the third-generation CD28-ζ-OX40 CAR, however, performed similar in modulating most CIK cell effector functions. Compared with the CD28-ζ CAR, however, the CD28-ζ-OX40 CAR accelerated terminal maturation of CD56(+) CIK cells producing high frequencies in activation-induced cell death (AICD) and reduced antitumor efficiency in vivo. Consequently, CD28-ζ CAR CIK cells of CD56(-) phenotype were superior in redirected tumor cell elimination. CAR-mediated CIK cell activation also increased antigen-independent target cell lysis; the CD28-ζ CAR was more efficient than the CD28-ζ-OX40 CAR. Translated into therapeutic strategies, CAR-redirected CIK cells benefit from CD28 costimulation; "super-costimulation" by the CD28-ζ-OX40 CAR, however, performed less in antitumor efficacy due to increased AICD.


Assuntos
Antígenos CD28/imunologia , Antígeno CD56/metabolismo , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/transplante , Neoplasias Experimentais/terapia , Receptores OX40/imunologia , Animais , Antígenos CD28/metabolismo , Antígeno CD56/imunologia , Morte Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Imunoterapia Adotiva , Ativação Linfocitária , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores OX40/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia
8.
Gastroenterology ; 143(4): 1095-107.e2, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22750462

RESUMO

BACKGROUND & AIMS: New treatment approaches are needed for patients with pancreatic adenocarcinoma. Carcinoembryonic antigen (CEA) is highly expressed on the surface of pancreatic adenocarcinoma cells; we investigated the effects of cytolytic T cells that recognize CEA in a mouse model of pancreatic carcinoma. METHODS: Immune-competent mice that expressed the CEA transgene (CEAtg) in the intestinal and pulmonary tracts were given intrapancreatic injections of Panc02 CEA(+) cells (express CEA and click beetle luciferase) and tumors were grown for 10 days. Mice were then given single intravenous injections of T cells engineered to express a chimeric antigen receptor (CAR) with high specificity, but moderate affinity, for CEA and a luminescence marker. RESULTS: Injection of the anti-CEA CAR T cells reduced the size of pancreatic tumors to below the limit of detection in all mice and produced long-term tumor eradication in 67% of mice. T cells also eradicated CEA(+) fibrosarcoma cells injected 45 days later. Bioluminescence imaging revealed the accumulation and persistence of the T cells at the tumor site. The efficacy of the T cells did not require lymphodepletion and was not reduced by soluble CEA. Mice developed some noninflammatory infiltrations of CAR(+) T cells in intestine and lung, but there was no evidence of destruction of CEA(+) healthy tissues. CONCLUSIONS: Injection of T cells that target CEA can eradicate tumors grown from CEA(+) pancreatic carcinoma cells in the pancreas of CEAtg mice without autoimmune effects.


Assuntos
Antígeno Carcinoembrionário/imunologia , Antígeno Carcinoembrionário/metabolismo , Carcinoma/terapia , Fibrossarcoma/terapia , Imunoterapia Adotiva , Neoplasias Pancreáticas/terapia , Linfócitos T Citotóxicos/transplante , Animais , Doenças Autoimunes/etiologia , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Carcinoma/metabolismo , Colite/imunologia , Fibrossarcoma/metabolismo , Imunoterapia Adotiva/efeitos adversos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Linfócitos T Citotóxicos/metabolismo
9.
Cells ; 12(2)2023 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-36672182

RESUMO

The efficacy of CD19-specific CAR T cells in the treatment of leukemia/lymphoma relies, at least in part, on the unique properties of the particular CAR and the presence of healthy B cells that enhance the target cell lysis and cytokine secretion through repetitive stimulation. Here, we report to apply the same CAR to target solid tumors, such as ErbB2+ carcinoma. CD19 CAR T cells are redirected towards the ErbB2+ cells by a fusion protein that is composed of the herceptin-derived anti-ErbB2 scFv 4D5 linked to the CD19 exodomain. The CD19-4D5scFv engager enabled CD19 CAR T cells to recognize the ErbB2+ cancer cells and to suppress the ErbB2+ tumor growth. The primary killing capacity by the ErbB2-redirected CD19 CAR T cells was as efficient as by the ErbB2 CAR T cells, however, adding CD19+ B cells furthermore reinforced the activation of the CD19 CAR T cells, thereby improving the anti-tumor activities. The ErbB2-redirected CD19 CAR T cells, moreover, showed a 100-fold superior selectivity in targeting cancer cells versus healthy fibroblasts, which was not the case for the ErbB2 CAR T cells. The data demonstrate that the CD19 CAR T cells can be high-jacked by a CD19-scFv engager protein to attack specifically solid cancer, thereby expanding their application beyond the B cell malignancies.


Assuntos
Neoplasias , Humanos , Neoplasias/terapia , Trastuzumab , Linfócitos B , Proteínas Adaptadoras de Transdução de Sinal , Linfócitos T , Receptor ErbB-2
10.
Front Immunol ; 14: 1110482, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36817444

RESUMO

In engineered T cells the CAR is co-expressed along with the physiological TCR/CD3 complex, both utilizing the same downstream signaling machinery for T cell activation. It is unresolved whether CAR-mediated T cell activation depends on the presence of the TCR and whether CAR and TCR mutually cross-activate upon engaging their respective antigen. Here we demonstrate that the CD3ζ CAR level was independent of the TCR associated CD3ζ and could not replace CD3ζ to rescue the TCR complex in CD3ζ KO T cells. Upon activation, the CAR did not induce phosphorylation of TCR associated CD3ζ and, vice versa, TCR activation did not induce CAR CD3ζ phosphorylation. Consequently, CAR and TCR did not cross-signal to trigger T cell effector functions. On the membrane level, TCR and CAR formed separate synapses upon antigen engagement as revealed by total internal reflection fluorescence (TIRF) and fast AiryScan microscopy. Upon engaging their respective antigen, however, CAR and TCR could co-operate in triggering effector functions through combinatorial signaling allowing logic "AND" gating in target recognition. Data also imply that tonic TCR signaling can support CAR-mediated T cell activation emphasizing the potential relevance of the endogenous TCR for maintaining T cell capacities in the long-term.


Assuntos
Receptores de Antígenos de Linfócitos T , Linfócitos T , Complexo CD3 , Complexo Receptor-CD3 de Antígeno de Linfócitos T/metabolismo , Transdução de Sinais , Receptores de Antígenos Quiméricos/imunologia
11.
Clin Dev Immunol ; 2012: 238924, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22481963

RESUMO

Adoptive therapy of malignant diseases with cytokine-induced killer (CIK) cells showed promise in a number of trials; the activation of CIK cells from cancer patients towards their autologous cancer cells still needs to be improved. Here, we generated CIK cells ex vivo from blood lymphocytes of colorectal cancer patients and engineered those cells with a chimeric antigen receptor (CAR) with an antibody-defined specificity for carcinoembryonic antigen (CEA). CIK cells thereby gained a new specificity as defined by the CAR and showed increase in activation towards CEA⁺ colon carcinoma cells, but less in presence of CEA⁻ cells, indicated by increased secretion of proinflammatory cytokines. Redirected CIK activation was superior by CAR-mediated CD28-CD3ζ than CD3ζ signaling only. CAR-engineered CIK cells from colon carcinoma patients showed improved activation against their autologous, primary carcinoma cells from biopsies resulting in more efficient tumour cell lysis. We assume that adoptive therapy with CAR-modified CIK cells shows improved selectivity in targeting autologous tumour lesions.


Assuntos
Carcinoma/imunologia , Neoplasias Colorretais/imunologia , Células Matadoras Induzidas por Citocinas/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Imunoterapia Adotiva/métodos , Antígenos CD28/imunologia , Complexo CD3/imunologia , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Carcinoma/patologia , Carcinoma/terapia , Engenharia Celular , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Células Matadoras Induzidas por Citocinas/efeitos dos fármacos , Células Matadoras Induzidas por Citocinas/patologia , Citocinas/imunologia , Citocinas/farmacologia , Humanos , Receptores de Antígenos/genética , Receptores de Antígenos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Células Tumorais Cultivadas
12.
Mol Ther ; 19(4): 760-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21326215

RESUMO

Adoptive T-cell transfer showed promising efficacy in recent trials raising interest in T cells with redirected specificity against tumors. T cells were engineered with a chimeric antigen receptor (CAR) with predefined binding and CD3ζ signaling to initiate T-cell activation. CD28 costimulation provided by a CD28-CD3ζ signaling CAR moreover improved T cell activation and persistence; however, it failed to meet the expectations with respect to mounting attacks against solid tumors infiltrated with regulatory T (Treg) cells. We revealed that a CD28 CAR-redirected T-cell attack is accompanied by higher numbers of Treg cells infiltrating the tumor and is less efficient against cancer cells in presence of Treg cells than a CD3ζ CAR T-cell attack. Deletion of the lck binding moiety in the CD28 CAR endodomain, however, improved redirected anti-tumor activity in presence of Treg cells without impairing interferon-γ (IFN-γ) secretion, proliferation, and cytolysis. CD28 modification abrogated interleukin-2 (IL-2) induction upon CAR engagement which in turn is no longer available to sustain Treg cell persistence. CARs with the modified CD28 endodomain thereby expedite the implementation of adoptive T-cell therapy in patients with a variety of cancer types that are heavily infiltrated by Treg cells.


Assuntos
Antígenos CD28/metabolismo , Interleucina-2/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD28/genética , Imuno-Histoquímica , Imunoterapia Adotiva , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-2/genética , Camundongos , Camundongos Nus , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T Reguladores/metabolismo
13.
Proc Natl Acad Sci U S A ; 106(14): 5784-8, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19307587

RESUMO

T-cell interaction with a target cell is a key event in the adaptive immune response and primarily driven by T-cell receptor (TCR) recognition of peptide-MHC (pMHC) complexes. TCR avidity for a given pMHC is determined by number of MHC molecules, availability of coreceptors, and TCR affinity for MHC or peptide, respectively, with peptide recognition being the most important factor to confer target specificity. Here we present high-resolution crystal structures of 2 Fab antibodies in complex with the immunodominant NY-ESO-1(157-165) peptide analogue (SLLMWITQV) presented by HLA-A*0201 and compare them with a TCR recognizing the same pMHC. Binding to the central methionine-tryptophan peptide motif and orientation of binding were almost identical for Fabs and TCR. As the MW "peg" dominates the contacts between Fab and peptide, we estimated the contributions of individual amino acids between the Fab and peptide to provide the rational basis for a peptide-focused second-generation, high-affinity antibody library. The final Fab candidate achieved better peptide binding by 2 light-chain mutations, giving a 20-fold affinity improvement to 2-4 nM, exceeding the affinity of the TCR by 1,000-fold. The high-affinity Fab when grafted as recombinant TCR on T cells conferred specific killing of HLA-A*0201/NY-ESO-1(157-165) target cells. In summary, we prove that affinity maturation of antibodies mimicking a TCR is possible and provide a strategy for engineering high-affinity antibodies that can be used in targeting specific pMHC complexes for diagnostic and therapeutic purposes.


Assuntos
Anticorpos/imunologia , Afinidade de Anticorpos/imunologia , Engenharia de Proteínas/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Afinidade de Anticorpos/genética , Cristalografia por Raios X , Citotoxicidade Imunológica , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Fragmentos Fab das Imunoglobulinas/imunologia , Mimetismo Molecular , Biblioteca de Peptídeos , Linfócitos T/imunologia
14.
Int J Cancer ; 129(12): 2935-44, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22030616

RESUMO

The therapeutic success of adoptive therapy with chimeric antigen receptor (CAR) engineered T cells depends on the appropriate costimulation of CD3ζ to induce full T cell activation. Costimulatory endodomains of the CD28 family are therefore fused with CD3ζ in a dual signalling CAR. Serious adverse events in two most recent trials; however, highlight the need to analyse in more detail the impact of each costimulatory endodomain on individual effector functions of redirected T cells. We therefore performed a thoroughly controlled side-by-side comparison of the most frequently used endodomains with respect to their impact on CD4(+) and CD8(+) T cell effector functions. CD28 reinforced T cell proliferation and is mandatory to induce IL-2. In the absence of added IL-2, CD28 and OX40 (CD137) but not 4-1BB (CD134) enhanced specific cytolysis. While CD28, 4-1BB and OX40 similarly improved pro-inflammatory cytokine secretion, OX40 most efficiently prevented activation induced cell death of CD62L(-) effector memory T cells. CD28 was superior to initiate the T cell response, OX40 and 4-1BB sustained the response in long term with OX40 being most effective. We consequently combined the beneficial functions in a 3rd generation CD28-OX40 CAR which substantially improved the antitumor response without loosing specificity.


Assuntos
Antígenos CD28/metabolismo , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores OX40/metabolismo , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , DNA Recombinante , Humanos , Ativação Linfocitária/imunologia , Camundongos , Transplante de Neoplasias , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia
15.
Clin Immunol ; 140(1): 71-83, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21482483

RESUMO

Sarcoidosis can evolve into a chronic disease with persistent granulomas accompanied by progressive fibrosis. While an unlimited inflammatory response suggests an impaired immune control in sarcoid lesions, it stands in contrast to the massive infiltration with CD4(+)CD25(high)FoxP3(+) regulatory T cells. We here revealed that those Treg cells in affected lung lesions were mainly derived from activated natural Treg cells with GARP (LRRC32)-positive phenotype but exhibited reduced repressor capacities despite high IL-10 and TGF-beta 1 levels. The repressive capacity of blood Treg cells, in contrast, was not impaired compared to age-matched healthy donors. Treg derived cells in granuloma lesions have undergone extensive rounds of amplifications indicated by shortened telomeres compared to blood Treg cells of the same patient. Lesional Treg derived cells moreover secreted pro-inflammatory cytokines including IL-4 which sustains granuloma formation through fibroblast amplification and the activation of mast cells, the latter indicated by the expression of membrane-bound oncostatin M.


Assuntos
Granuloma/imunologia , Sarcoidose Pulmonar/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Proliferação de Células , Separação Celular , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Memória Imunológica/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
16.
Cells ; 9(4)2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32260097

RESUMO

Chimeric antigen receptor (CAR) redirected T cells are efficacious in the treatment of leukemia/lymphoma, however, showed less capacities in eliminating solid tumors which is thought to be partly due to the lack of cytokine support in the tumor lesion. In order to deliver supportive cytokines, we took advantage of the inherent ability of mesenchymal stem cells (MSCs) to actively migrate to tumor sites and engineered MSCs to release both IL7 and IL12 to promote homeostatic expansion and Th1 polarization. There is a mutual interaction between engineered MSCs and CAR T cells; in presence of CAR T cell released IFN-γ and TNF-α, chronic inflammatory Th2 MSCs shifted towards a Th17/Th1 pattern with IL2 and IL15 release that mutually activated CAR T cells with extended persistence, amplification, killing and protection from activation induced cell death. MSCs releasing IL7 and IL12 were superior over non-modified MSCs in supporting the CAR T cell response and improved the anti-tumor attack in a transplant tumor model. Data demonstrate the first use of genetically modified MSCs as vehicles to deliver immuno-modulatory proteins to the tumor tissue in order to improve the efficacy of CAR T cells in the treatment of solid malignancies.


Assuntos
Neoplasias Colorretais/imunologia , Interleucina-12/metabolismo , Interleucina-7/metabolismo , Células-Tronco Mesenquimais/metabolismo , Engenharia de Proteínas , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Animais , Antígenos/imunologia , Proliferação de Células , Sobrevivência Celular , Citocinas/metabolismo , Células HEK293 , Humanos , Camundongos , Transplante de Neoplasias
17.
Rejuvenation Res ; 11(5): 915-33, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18803477

RESUMO

Repetitive antigen encounters together with a strong CD28 co-stimulatory signal were recently identified as driving extensive amplification of human regulatory T (Treg) cells; however, the consequences of this on the functional capacities of Treg cells remain unknown. In this report, we reveal that T cell receptor (TCR)/CD28-triggered amplification in vitro converts CD4+CD25(high)FoxP3+ Treg cells into a late memory phenotype associated with immunosenescence and loss of CD7. Accordingly, ex vivo-isolated CD7- Treg cells have shortened telomeres and decreased telomerase expression compared to the majority of "mature" CD7+ Treg cells. Although they resist spontaneous apoptosis, amplified CD7- Treg cells exhibit increased sensitivity to activation-induced cell death (AICD). Extensive amplification of Treg cells is, moreover, accompanied by an increased activation threshold, reduced suppressor capacities, and interleukin-10 (IL-10) secretion, but secretion of high amounts of IL-4. Concomitantly, amplified Treg cells express homing receptors targeting them to the periphery. This is confirmed in vivo by the extensive accumulation of CD7- Treg cells with shortened telomeres in chronic inflammatory skin lesions, including atopic dermatitis and lichen ruber. Our data indicate that extensive amplification upon repetitive TCR/CD28 engagement alters the functional capacities of CD4+CD25(high) Treg cells toward less suppressive cells, but potential mediators in sustaining inflammatory reactions through IL-4.


Assuntos
Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/fisiologia , Adulto , Antígenos CD7/metabolismo , Diferenciação Celular , Proliferação de Células , Senescência Celular , Citocinas/biossíntese , Dermatite/imunologia , Dermatite/patologia , Humanos , Memória Imunológica , Técnicas In Vitro , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/fisiologia , Linfócitos T Reguladores/citologia , Adulto Jovem
18.
Cancers (Basel) ; 9(9)2017 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-28850063

RESUMO

Evidences are accumulating that CD4⁺ T cells can physiologically mediate antigen specific target cell lysis. By circumventing major histocompatibility complex (MHC)-restrictions through an engineered chimeric antigen receptor (CAR), CD4⁺ T cells lyse defined target cells as efficiently as do CD8⁺ T cells. However, the cytolytic capacity of redirected CD4⁺CD25- T cells, in comparison with CD4⁺CD25⁺ regulatory T (Treg) cells was so far not thoroughly defined. Treg cells require a strong CD28 signal together with CD3ζ for activation. We consequently used a CAR with combined CD28-CD3ζ signalling for redirecting CD4⁺CD25- T cells and CD4⁺CD25⁺ Treg cells from the same donor. CAR redirected activation of these T cell subsets and induced a distinct cytokine pattern with high IL-10 and a lack of IL-2 release by Treg cells. Despite strong antigen-specific activation, CAR Treg cells produced only weak target cell lysis, whereas CD4⁺CD25- CAR T cells were potent killers. Cytolysis did not correlate with the target cell sensitivity to Fas/FasL mediated killing; CD4⁺CD25- T cells upregulated perforin and granzyme B upon CAR activation, whereas Treg cells did less. The different cytolytic capacities of CAR redirected conventional CD4⁺ cells and Treg cells imply their use for different purposes in cell therapy.

19.
Expert Rev Clin Immunol ; 13(2): 151-155, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27546707

RESUMO

INTRODUCTION: Adoptive therapy with chimeric antigen receptor (CAR) T cells redirected towards CD19 produces remissions of B cell malignancies, however, it also eradicates healthy B cells sharing the target antigen. Such 'on-target off-tumor' toxicity raises serious safety concerns when the target antigen is also expressed by tissue stem cells, with the risk of lasting tissue destruction. Areas covered: We discuss CAR T cell targeting of activation antigens versus lineage associated antigens on the basis of recent experimental and animal data and the literature in the field. Expert commentary: Targeting an activation associated antigen which is transiently expressed by stem cells seems to be safe, like CAR T cells targeting CD30 spare CD30+ hematopoietic stem and progenitor cells while eliminating CD30+ lymphoma cells, whereas targeting lineage associated antigens which increase in expression during cell maturation, like folate receptor-ß and CD123, is of risk to destruct tissue stem cells.


Assuntos
Células-Tronco Adultas/metabolismo , Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/fisiologia , Células-Tronco Adultas/imunologia , Animais , Autoimunidade , Humanos , Imunoterapia Adotiva/tendências , Mimetismo Molecular , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T/transplante
20.
Hum Gene Ther ; 28(12): 1147-1157, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29207878

RESUMO

The treatment of leukemia/lymphoma by chimeric antigen receptor (CAR) redirected T cells with specificity for CD19 induced complete remissions in the majority of patients, with a realistic hope for cure. However, recent follow-up data revealed a substantial risk of relapse through leukemic cells that lack the CAR targeted antigen. In this situation, a bispecific CAR with binding domains for CD19 and CD20 is aimed at recognizing leukemic cells with only one cognate antigen. The anti-CD20-CD19 bispecific CAR induced a full T-cell response upon engagement of CD19 or CD20 on target cells showing a true "OR" gate recognition in redirecting T-cell activation. T cells with the anti-CD20-CD19 CAR efficiently killed patients' chronic lymphocytic leukemia cells in vitro. The bispecific CAR T cells cleared pediatric acute lymphocytic leukemia with a mixed CD19+CD20+/CD20- phenotype from the blood and bone marrow of transplanted mice, while anti-CD20 CAR T cells left CD20- leukemic cells behind without curing the disease. Data indicate the superior anti-leukemic activity in the control of leukemia, implying that the anti-CD20-CD19 bispecific CAR T cells may reduce the risk of relapse through antigen-loss leukemic cells in the long term.


Assuntos
Antígenos CD19/imunologia , Antígenos CD20/imunologia , Imunoterapia Adotiva/métodos , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Receptores de Antígenos de Linfócitos T , Proteínas Recombinantes de Fusão , Linfócitos T , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Linfoma de Células B/genética , Masculino , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA