Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Blood ; 143(6): 507-521, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38048594

RESUMO

ABSTRACT: Chimeric antigen receptor T-cell (CAR T) therapy has produced remarkable clinical responses in B-cell neoplasms. However, many challenges limit this class of agents for the treatment of other cancer types, in particular the lack of tumor-selective antigens for solid tumors and other hematological malignancies, such as acute myeloid leukemia (AML), which may be addressed without significant risk of severe toxicities while providing sufficient abundance for efficient tumor suppression. One approach to overcome this hurdle is dual targeting by an antibody-T-cell receptor (AbTCR) and a chimeric costimulatory signaling receptor (CSR) to 2 different antigens, in which both antigens are found together on the cancer cells but not together on normal cells. To explore this proof of concept in AML, we engineered a new T-cell format targeting Wilms tumor 1 protein (WT1) and CD33; both are highly expressed on most AML cells. Using an AbTCR comprising a newly developed TCR-mimic monoclonal antibody against the WT1 RMFPNAPYL (RMF) epitope/HLA-A2 complex, ESK2, and a secondary CSR comprising a single-chain variable fragment directed to CD33 linked to a truncated CD28 costimulatory fragment, this unique platform confers specific T-cell cytotoxicity to the AML cells while sparing healthy hematopoietic cells, including CD33+ myelomonocytic normal cells. These data suggest that this new platform, named AbTCR-CSR, through the combination of a AbTCR CAR and CSR could be an effective strategy to reduce toxicity and improve specificity and clinical outcomes in adoptive T-cell therapy in AML.


Assuntos
Leucemia Mieloide Aguda , Anticorpos de Cadeia Única , Humanos , Linfócitos T , Receptores de Antígenos de Linfócitos T , Leucemia Mieloide Aguda/patologia , Imunoterapia Adotiva
2.
Cancer Immunol Immunother ; 72(11): 3773-3786, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37635172

RESUMO

Epithelial ovarian cancer is the most lethal of gynecological cancers. The therapeutic efficacy of chimeric antigen receptor (CAR) T cell directed against single antigens is limited by the heterogeneous target antigen expression in epithelial ovarian tumors. To overcome this limitation, we describe an engineered cell with both dual targeting and orthogonal cytotoxic modalities directed against two tumor antigens that are highly expressed on ovarian cancer cells: cell surface Muc16 and intracellular WT1. Muc16-specific CAR T cells (4H11) were engineered to secrete a bispecific T cell engager (BiTE) constructed from a TCR mimic antibody (ESK1) reactive with the WT1-derived epitope RMFPNAPYL (RMF) presented by HLA-A2 molecules. The secreted ESK1 BiTE recruited and redirected other T cells to WT1 on the tumor cells. We show that ESK1 BiTE-secreting 4H11 CAR T cells exhibited enhanced anticancer activity against cancer cells with low Muc16 expression, compared to 4H11 CAR T cells alone, both in vitro and in mouse tumor models. Dual orthogonal cytotoxic modalities with different specificities targeting both surface and intracellular tumor-associated antigens present a promising strategy to overcome resistance to CAR T cell therapy in epithelial ovarian cancer and other cancers.


Assuntos
Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Humanos , Camundongos , Feminino , Animais , Carcinoma Epitelial do Ovário/terapia , Neoplasias Ovarianas/terapia , Antígenos de Neoplasias , Linfócitos T , Proteínas WT1
3.
Cancer Immunol Immunother ; 70(5): 1189-1202, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33123756

RESUMO

Identification of neoepitopes as tumor-specific targets remains challenging, especially for cancers with low mutational burden, such as ovarian cancer. To identify mutated human leukocyte antigen (HLA) ligands as potential targets for immunotherapy in ovarian cancer, we combined mass spectrometry analysis of the major histocompatibility complex (MHC) class I peptidomes of ovarian cancer cells with parallel sequencing of whole exome and RNA in a patient with high-grade serous ovarian cancer. Four of six predicted mutated epitopes capable of binding to HLA-A*02:01 induced peptide-specific T cell responses in blood from healthy donors. In contrast, all six peptides failed to induce autologous peptide-specific response by T cells in peripheral blood or tumor-infiltrating lymphocytes from ascites of the patient. Surprisingly, T cell responses against a low-affinity p53-mutant Y220C epitope were consistently detected in the patient with either unprimed or in vitro peptide-stimulated T cells even though the patient's primary tumor did not bear this mutation. Our results demonstrated that tumor heterogeneity and distinct immune microenvironments within a patient should be taken into consideration for identification of immunogenic neoantigens. T cell responses to a driver gene-derived p53 Y220C mutation in ovarian cancer warrant further study.


Assuntos
Antígenos de Neoplasias/metabolismo , Epitopos de Linfócito T/metabolismo , Antígeno HLA-A2/metabolismo , Imunoterapia Adotiva/métodos , Mutação/genética , Neoplasias Ovarianas/imunologia , Linfócitos T/imunologia , Proteína Supressora de Tumor p53/metabolismo , Antígenos de Neoplasias/genética , Células Cultivadas , Epitopos de Linfócito T/genética , Feminino , Antígeno HLA-A2/genética , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Microambiente Tumoral , Proteína Supressora de Tumor p53/genética , Sequenciamento do Exoma
4.
Res Sq ; 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37214945

RESUMO

Epithelial ovarian cancer is the most lethal of gynecological cancers. The therapeutic efficacy of chimeric antigen receptor (CAR) T cell directed against single antigens is limited by the heterogeneous target antigen expression in epithelial ovarian tumors. To overcome this limitation, we describe an engineered cell with both dual targeting and orthogonal cytotoxic modalities directed against two tumor antigens that are highly expressed on ovarian cancer cells: cell surface Muc16 and intracellular WT1. Muc16-specific CAR-T cells (4H11) were engineered to secrete a bispecific T cell engager (BiTE) constructed from a TCR mimic antibody (ESK1) reactive with the WT1-derived epitope RMFPNAPYL (RMF) presented by HLA-A2 molecules. The secreted ESK1 BiTE recruited and redirected other T cells to WT1 on the tumor cells. We show that ESK1 BiTE-secreting 4H11 CAR-T cells exhibited enhanced anticancer activity against cancer cells with low Muc16 expression, compared to 4H11 CAR-T cells alone, both in vitro and in mouse tumor models. Dual orthogonal cytotoxic modalities with different specificities targeting both surface and intracellular tumor-associated antigens present a promising strategy to overcome resistance to CAR-T cell therapy in epithelial ovarian cancer and other cancers.

5.
Blood ; 116(2): 171-9, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20400682

RESUMO

A pilot study was undertaken to assess the safety, activity, and immunogenicity of a polyvalent Wilms tumor gene 1 (WT1) peptide vaccine in patients with acute myeloid leukemia in complete remission but with molecular evidence of WT1 transcript. Patients received 6 vaccinations with 4 WT1 peptides (200 microg each) plus immune adjuvants over 12 weeks. Immune responses were evaluated by delayed-type hypersensitivity, CD4+ T-cell proliferation, CD3+ T-cell interferon-gamma release, and WT1 peptide tetramer staining. Of the 9 evaluable patients, 7 completed 6 vaccinations and WT1-specific T-cell responses were noted in 7 of 8 patients. Three patients who were HLA-A0201-positive showed significant increase in interferon-gamma-secreting cells and frequency of WT1 tetramer-positive CD8+ T cells. Three patients developed a delayed hypersensitivity reaction after vaccination. Definite related toxicities were minimal. With a mean follow-up of 30 plus or minus 8 months after diagnosis, median disease-free survival has not been reached. These preliminary data suggest that this polyvalent WT1 peptide vaccine can be administered safely to patients with a resulting immune response. Further studies are needed to establish the role of vaccination as viable postremission therapy for acute myeloid leukemia.


Assuntos
Vacinas Anticâncer/uso terapêutico , Leucemia Mieloide Aguda/terapia , Proteínas Oncogênicas/uso terapêutico , Vacinação/métodos , Proteínas WT1/uso terapêutico , Adulto , Idoso , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Citotoxicidade Imunológica , Intervalo Livre de Doença , Feminino , Antígenos HLA-A/genética , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Hipersensibilidade Tardia/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/imunologia , Projetos Piloto , Indução de Remissão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Proteínas WT1/imunologia , Adulto Jovem
6.
PLoS One ; 17(3): e0265534, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35298559

RESUMO

More effective treatments are needed for human papilloma virus (HPV)-induced cancers despite HPV virus vaccination. The oncogenic HPV protein targets are currently undruggable and intracellular and therefore there are no antibodies to these targets. Here we report the discovery of TCR mimic monoclonal antibodies (TCRm mAb) specific for the HPV E7 protein p11-19, YMLDLQPET, when presented on the cell surface in the context of HLA-A*02:01 by use of human phage display libraries. One of the mAbs, 3F8, was able to specifically mediate T cell- redirected cytotoxicity, in a bispecific T cell engager (BiTE) form. While further studies are required to assess the therapeutic potential of this approach, the study provided the proof of concept that TCRm mAb could be a therapeutic strategy for HPV-induced human cancers.


Assuntos
Antineoplásicos Imunológicos , Neoplasias , Proteínas Oncogênicas Virais , Infecções por Papillomavirus , Anticorpos Monoclonais , Epitopos , Antígenos HLA-A , Papillomavirus Humano 16 , Humanos , Neoplasias/tratamento farmacológico , Proteínas E7 de Papillomavirus , Infecções por Papillomavirus/tratamento farmacológico , Receptores de Antígenos de Linfócitos T
7.
JCI Insight ; 7(5)2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260532

RESUMO

Phosphopeptides derived from dysregulated protein phosphorylation in cancer cells can be processed and presented by MHC class I and class II molecules and, therefore, represent an untapped class of tumor-specific antigens that could be used as widely expressed "public" cancer neoantigens (NeoAgs). We generated a TCR mimic (TCRm) mAb, 6B1, specific for a phosphopeptide derived from insulin receptor substrate 2 (pIRS2) presented by HLA-A*02:01. The pIRS2 epitope's presentation by HLA-A*02:01 was confirmed by mass spectrometry. The TCRm 6B1 specifically bound to pIRS2/HLA-A2 complex on tumor cell lines that expressed pIRS2 in the context of HLA-A*02:01. Bispecific mAbs engaging CD3 of T cells were able to kill tumor cell lines in a pIRS2- and HLA-A*02:01-restricted manner. Structure modeling shows a prerequisite for an arginine or lysine at the first position to bind mAb. Therefore, 6B1 could recognize phosphopeptides derived from various phosphorylated proteins with similar amino acid compositions. This raised the possibility that a TCRm specific for the pIRS2/HLA-A2 complex could target a range of phosphopeptides presented by HLA-A*02:01 in various tumor cells. This is the first TCRm mAb to our knowledge targeting a phosphopeptide/MHC class I complex; the potential of this class of agents for clinical applications warrants further investigation.


Assuntos
Antígeno HLA-A2 , Fosfopeptídeos , Anticorpos Monoclonais/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Fosfopeptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
8.
Cancer Immunol Immunother ; 59(10): 1467-79, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20532500

RESUMO

BACKGROUND: The transcription factor, WT1, is highly overexpressed in malignant pleural mesothelioma (MPM) and immunohistochemical stains for WT1 are used routinely to aid in its diagnosis. Using computer prediction analysis we designed analog peptides derived from WT1 sequences by substituting amino acids at key HLA-A0201 binding positions. We tested the safety and immunogenicity of a WT1 vaccine comprised of four class I and class II peptides in patients with thoracic neoplasms expressing WT1. METHODS: Therapy consisted of six subcutaneous vaccinations administered with Montanide adjuvant on weeks 0, 4, 6, 8, 10, and 12, with 6 additional monthly injections for responding patients. Injection sites were pre-stimulated with GM-CSF (70 mcg). Immune responses were evaluated by DTH, CD4 T-cell proliferation, CD8 T-cell interferon gamma release, intracellular cytokine staining, WT1 peptide MHC-tetramer staining, and cytotoxicity against WT1 positive tumor cells. RESULTS: Nine patients with MPM and 3 with NSCLC were vaccinated, with 8 patients receiving at least 6 vaccinations; in total, 10 patients were evaluable for immune response. Six out of nine patients tested demonstrated CD4 T-cell proliferation to WT1 specific peptides, and five of the six HLA-A0201 patients tested mounted a CD8 T-cell response. Stimulated T cells were capable of cytotoxicity against WT-1 positive cells. Vaccination also induced polyfunctional CD8 T cell responses. CONCLUSIONS: This multivalent WT1 peptide analog vaccine induces immune responses in a high proportion of patients with thoracic malignancies with minimal toxicity. A randomized trial testing this vaccine as adjuvant therapy in MPM is planned.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas , Mesotelioma , Fragmentos de Peptídeos , Proteínas WT1/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular , Feminino , Humanos , Imuno-Histoquímica , Imunoterapia , Masculino , Mesotelioma/imunologia , Mesotelioma/terapia , Pessoa de Meia-Idade , Dados de Sequência Molecular , Estadiamento de Neoplasias , Fragmentos de Peptídeos/genética , Proteínas WT1/administração & dosagem , Proteínas WT1/genética
9.
Cancer Biother Radiopharm ; 35(6): 459-473, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32013538

RESUMO

An α particle-emitting nanodrug that is a potent and specific antitumor agent and also prompts significant remodeling of local immunity in the tumor microenvironment (TME) has been developed and may impact the treatment of melanoma. Biocompatible ultrasmall fluorescent core-shell silica nanoparticles (C' dots, diameter ∼6.0 nm) have been engineered to target the melanocortin-1 receptor expressed on melanoma through α melanocyte-stimulating hormone peptides attached to the C' dot surface. Actinium-225 is also bound to the nanoparticle to deliver a densely ionizing dose of high-energy α particles to cancer. Nanodrug pharmacokinetic properties are optimal for targeted radionuclide therapy as they exhibit rapid blood clearance, tumor-specific accumulation, minimal off-target localization, and renal elimination. Potent and specific tumor control, arising from the α particles, was observed in a syngeneic animal model of melanoma. Surprisingly, the C' dot component of this drug initiates a favorable pseudopathogenic response in the TME generating distinct changes in the fractions of naive and activated CD8 T cells, Th1 and regulatory T cells, immature dendritic cells, monocytes, MΦ and M1 macrophages, and activated natural killer cells. Concomitant upregulation of the inflammatory cytokine genome and adaptive immune pathways each describes a macrophage-initiated pseudoresponse to a viral-shaped pathogen. This study suggests that therapeutic α-particle irradiation of melanoma using ultrasmall functionalized core-shell silica nanoparticles potently kills tumor cells, and at the same time initiates a distinct immune response in the TME.


Assuntos
Partículas alfa/uso terapêutico , Portadores de Fármacos/química , Melanoma Experimental/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Neoplasias Cutâneas/radioterapia , Microambiente Tumoral/efeitos da radiação , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Linhagem Celular Tumoral/transplante , Biologia Computacional , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Imunidade Celular/genética , Imunidade Celular/efeitos da radiação , Masculino , Dose Máxima Tolerável , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Terapia de Alvo Molecular/métodos , Nanopartículas/química , RNA-Seq , Compostos Radiofarmacêuticos/farmacocinética , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , Receptor Tipo 1 de Melanocortina/metabolismo , Dióxido de Silício/química , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Distribuição Tecidual , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
10.
Cancer Immunol Res ; 7(12): 1984-1997, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31540894

RESUMO

T-cell immunotherapies are often thwarted by the limited presentation of tumor-specific antigens abetted by the downregulation of human leukocyte antigen (HLA). We showed that drugs inhibiting ALK and RET produced dose-related increases in cell-surface HLA in tumor cells bearing these mutated kinases in vitro and in vivo, as well as elevated transcript and protein expression of HLA and other antigen-processing machinery. Subsequent analysis of HLA-presented peptides after ALK and RET inhibitor treatment identified large changes in the immunopeptidome with the appearance of hundreds of new antigens, including T-cell epitopes associated with impaired peptide processing (TEIPP) peptides. ALK inhibition additionally decreased PD-L1 levels by 75%. Therefore, these oncogenes may enhance cancer formation by allowing tumors to evade the immune system by downregulating HLA expression. Altogether, RET and ALK inhibitors could enhance T-cell-based immunotherapies by upregulating HLA, decreasing checkpoint blockade ligands, and revealing new, immunogenic, cancer-associated antigens.


Assuntos
Quinase do Linfoma Anaplásico/antagonistas & inibidores , Antígenos de Neoplasias/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-ret/antagonistas & inibidores , Animais , Apresentação de Antígeno/efeitos dos fármacos , Linhagem Celular Tumoral , Crizotinibe/farmacologia , Feminino , Humanos , Camundongos Transgênicos , Neoplasias/imunologia , Peptídeos/imunologia , Pirimidinas/farmacologia , Sulfonas/farmacologia
11.
Oncoimmunology ; 8(7): 1570778, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31143508

RESUMO

Depletion of T regulatory cells (Tregs) in the tumor microenvironment is a promising cancer immunotherapy strategy. Current approaches for depleting Tregs are limited by lack of specificity and concurrent depletion of anti-tumor effector T cells. The transcription factor forkhead box p3 (Foxp3) plays a central role in the development and function of Tregs and is an ideal target in Tregs, but Foxp3 is an intracellular, undruggable protein to date. We have generated a T cell receptor mimic antibody, "Foxp3-#32," recognizing a Foxp3-derived epitope in the context of HLA-A*02:01. The mAb Foxp3-#32 selectively recognizes CD4 + CD25 + CD127low and Foxp3 + Tregs also expressing HLA-A*02:01 and depletes these cells via antibody-mediated cellular cytotoxicity. Foxp3-#32 mAb depleted Tregs in xenografts of PBMCs from a healthy donor and ascites fluid from a cancer patient. A TCRm mAb targeting intracellular Foxp3 epitope represents an approach to deplete Tregs.

12.
Clin Cancer Res ; 13(15 Pt 1): 4547-55, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671141

RESUMO

PURPOSE: Wilms' tumor 1 protein (WT1), a transcription factor overexpressed in malignant mesothelioma, leukemias, and other solid tumors, is an ideal target for immunotherapy. WT1 class I peptide epitopes that were identified and shown to stimulate CD8(+) T cells are being tested as vaccine candidates in several clinical trials. The induction and maintenance of a robust memory CD8(+) cytotoxic T-cell response requires CD4(+) T-cell help. EXPERIMENTAL DESIGN: Three HLA class II peptide epitopes of WT1 with high predictive affinities to multiple HLA-DRB1 molecules were identified using the SYFPEITHI algorithm. Due to the highly polymorphic nature of the HLA class II alleles, such reactivity is critical in the development of a broadly useful therapeutic. One of the WT1 CD4(+) peptide epitopes, 122-140, comprises a previously identified CD8(+) peptide epitope (126-134). By mutating residue 126 from an arginine to a tyrosine, we embedded a synthetic immunogenic analogue CD8(+) epitope (126-134) inside the longer peptide (122-140). This analogue was previously designed to improve immunogenicity and induce a potent CD8(+) response. RESULTS: WT1 peptides 328-349 and 423-441 are able to stimulate a peptide-specific CD4(+) response that can recognize WT1(+) tumor cells in multiple HLA-DRB1 settings as determined by IFN-gamma enzyme-linked immunospot assays. The mutated WT1 peptide epitope 122-140 is able to induce CD4(+) and cytotoxic CD8(+) WT1-specific T-cell responses that can recognize the native WT1 epitopes on the surface of human WT1(+) cancer cells. Cross-priming experiments showed that antigen-presenting cells pulsed with either mesothelioma or leukemia tumor lysates can process and present each of the CD4(+) peptides identified. CONCLUSIONS: These studies provide the rationale for using the WT1 CD4(+) peptides in conjunction with CD8(+) peptide epitopes to vaccinate patients with WT1-expressing cancers.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos/imunologia , Mesotelioma/patologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Proteínas WT1/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Cromo/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Epitopos/genética , Antígenos HLA-DR/metabolismo , Cadeias HLA-DRB1 , Humanos , Imunização , Interferon gama/metabolismo , Leucemia/imunologia , Leucemia/patologia , Leucemia/terapia , Mesotelioma/imunologia , Mesotelioma/terapia , Fragmentos de Peptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/patologia , Proteínas WT1/genética , Proteínas WT1/metabolismo
13.
Oncoimmunology ; 6(2): e1252895, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28344864

RESUMO

The Wilms' tumor oncogene protein (WT1) is a highly validated tumor antigen for immunotherapy. WT1-targeted immunotherapy has been extensively explored in multiple human trials in various cancers. However, clinical investigations using WT1 epitopes have generally focused on two peptides, HLA-restricted to HLA-A*02:01 or HLA-A*24:02. The goal of this study was to identify new epitopes derived from WT1, to expand the potential use of WT1 as a target of immunotherapy. Using computer-based MHC-binding algorithms and in vitro validation of the T cell responses specific for the identified peptides, we found that a recently identified HLA-A*24:02-binding epitope (239-247), NQMNLGATL (NQM), was also a strong CD8+ T cell epitope for HLA-A*02:01 molecule. A peptide second position Q240L substitution (NLM) or Q240Y substitution (NYM), further enhanced the T cell responses in both HLA-A*02:01 positive and HLA-A*24:02 positive healthy donors. Importantly, T cells stimulated with the new analog peptides displayed heteroclitic cross-reactivity with the native NQM sequence and were able to kill HLA-matched WT1-positive tumor cell lines and primary leukemia blasts. In addition, longer native and heteroclitic HLA-DR.B1-binding peptides, comprising the nine amino acid NQM or NLM sequences, could induce T cell response that recognized the CD8+ epitope NQM, suggesting the processing and the presentation by HLA-A*02:01 molecules of the CD8+ T cell epitope embedded within it. Our studies suggest that the analog peptides NLM and NYM could be potential candidates for future immunotherapy targeting WT1 positive cancers in the context of HLA-A*02:01 and A*24:02 positive populations.

14.
J Clin Invest ; 127(7): 2705-2718, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28628042

RESUMO

Preferentially expressed antigen in melanoma (PRAME) is a cancer-testis antigen that is expressed in many cancers and leukemias. In healthy tissue, PRAME expression is limited to the testes and ovaries, making it a highly attractive cancer target. PRAME is an intracellular protein that cannot currently be drugged. After proteasomal processing, the PRAME300-309 peptide ALYVDSLFFL (ALY) is presented in the context of human leukocyte antigen HLA-A*02:01 molecules for recognition by the T cell receptor (TCR) of cytotoxic T cells. Here, we have described Pr20, a TCR mimic (TCRm) human IgG1 antibody that recognizes the cell-surface ALY peptide/HLA-A2 complex. Pr20 is an immunological tool and potential therapeutic agent. Pr20 bound to PRAME+HLA-A2+ cancers. An afucosylated Fc form (Pr20M) directed antibody-dependent cellular cytotoxicity against PRAME+HLA-A2+ leukemia cells and was therapeutically effective against mouse xenograft models of human leukemia. In some tumors, Pr20 binding markedly increased upon IFN-γ treatment, mediated by induction of the immunoproteasome catalytic subunit ß5i. The immunoproteasome reduced internal destructive cleavages within the ALY epitope compared with the constitutive proteasome. The data provide rationale for developing TCRm antibodies as therapeutic agents for cancer, offer mechanistic insight on proteasomal regulation of tumor-associated peptide/HLA antigen complexes, and yield possible therapeutic solutions to target antigens with ultra-low surface presentation.


Assuntos
Antígenos de Neoplasias/imunologia , Antígeno HLA-A1/imunologia , Imunoglobulina G/farmacologia , Neoplasias Experimentais , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Imunoglobulina G/imunologia , Masculino , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Clin Cancer Res ; 11(24 Pt 1): 8763-72, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16361564

RESUMO

PURPOSE: Presentation of exogenous antigen by MHC class I molecules, or cross-presentation, is a property of dendritic cells, which is considered crucial for the priming of cytotoxic T-cell response to tumor antigens. However, the precise mechanisms of this process are not fully understood. EXPERIMENTAL DESIGN AND RESULTS: We show here in a human in vitro system, using B lymphoma cells as a tumor model, that the cross-presentation of cell-associated antigens to T cells by dendritic cells requires "help" from natural killer cells. When autologous dendritic cells that had taken up apoptotic B lymphoma cells and induced to a fully mature state were used to stimulate nonadherent cells of peripheral blood mononuclear cells from healthy donors, they induced strong cytotoxicity against B lymphoma cells in a HLA-A0201-restricted manner. The cells failed to induce cytotoxicity, however, when purified T cells were used as effector cells. Depletion of CD56+ cells, but not CD14+ or CD19+ cells, abrogated the cytotoxicity of nonadherent cells, showing that the help was provided by natural killer cells. Further, when natural killer cells were present in the cultures, a strong and persistent production of interleukin-18, but not interleukin-12 and interleukin-15, was observed. Blocking interleukin-18 significantly reduced the cytotoxicity of nonadherent cells against B lymphoma cells. CONCLUSIONS: These results suggest that capture of tumor cells and a full maturation status of dendritic cells are not sufficient to cross-prime CD8 T cells. Effective cross-priming requires further activation of dendritic cells by natural killer cells and an abundant production of interleukin-18, which, along with other yet undefined mechanisms, contribute to the generation of CTL response against B-cell lymphoma.


Assuntos
Antígenos de Neoplasias/imunologia , Apresentação Cruzada/imunologia , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Linfoma de Células B/imunologia , Anticorpos Bloqueadores/farmacologia , Antígenos de Neoplasias/análise , Antígeno CD56/análise , Antígeno CD56/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Interleucina-18/antagonistas & inibidores , Interleucina-18/metabolismo , Depleção Linfocítica , Fagocitose , Linfócitos T Citotóxicos/imunologia
16.
Haematologica ; 90(10): 1324-32, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16219568

RESUMO

BACKGROUND AND OBJECTIVES: Chronic myelogenous leukemia (CML) presents a unique opportunity to develop therapeutic strategies using vaccination against a truly tumor-specific antigen that is also the oncogenic protein required for neoplasia. We have shown in phase I and II trials that a tumor-specific, bcr-abl-derived peptide vaccine can be safely administered to patients with chronic phase CML and can elicit a reliable specific CD4 immune response. However, variable CD8 responses and no HLA A0201-restricted responses were found. One strategy to circumvent this poor immunogenicity is to design synthetic immunogenic analog peptides that cross-react with the native peptides (a heteroclitic response). The aim of this study was to design such peptides. DESIGN AND METHODS: By using computer prediction analysis. We designed a number of synthetic peptides derived from the junctional sequences of CML (p210/b3a2 or p210/b2a2) in which single and double amino acid substitutions were introduced at key HLA A0201 binding positions. The binding of these peptides was tested by a thermostabilization assay using a T2 cell line. RESULTS: We found three peptides that predicted good binding to HLA A0201 molecules and stabilized MHC class I A0201 molecules on the surface of T2 cell lines. These peptides were screened for eliciting HLA restricted, peptide-specific cytotoxic T lymphocyte responses using CD3+ T cells from several A0201 donors and CML patients. The CD8+ cytotoxic T lymphocytes lines were assessed by either interferon-g ELISPOT or a chromium release assay using pulsed, HLA-matched leukemic cell lines. The analog peptides generated larger immune responses (increased CD8 T-cell precursor frequency) than did the native peptides. Importantly, CD8+ T cells stimulated with the new synthetic peptides cross-reacted with the native bcr-abl peptides. INTERPRETATION AND CONCLUSIONS: In conclusion, analog CML fusion peptides with increased immunogenicity and heteroclitic properties can be synthesized and may be useful in vaccination strategies.


Assuntos
Proteínas de Fusão bcr-abl/química , Proteínas de Fusão bcr-abl/fisiologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Linhagem Celular Tumoral , Técnicas de Química Combinatória/métodos , Antígenos HLA-A/imunologia , Antígenos HLA-A/metabolismo , Humanos , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/imunologia , Linfócitos T/metabolismo
17.
Nat Biotechnol ; 33(10): 1079-86, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26389576

RESUMO

Intracellular tumor antigens presented on the cell surface in the context of human leukocyte antigen (HLA) molecules have been targeted by T cell-based therapies, but there has been little progress in developing small-molecule drugs or antibodies directed to these antigens. Here we describe a bispecific T-cell engager (BiTE) antibody derived from a T-cell receptor (TCR)-mimic monoclonal antibody (mAb) ESK1, which binds a peptide derived from the intracellular oncoprotein WT1 presented on HLA-A*02:01. Despite the very low density of the complexes at the cell surface, ESK1-BiTE selectively activated and induced proliferation of cytolytic human T cells that killed cells from multiple leukemias and solid tumors in vitro and in mice. We also discovered that in an autologous in vitro setting, ESK1-BiTE induced a robust secondary CD8 T-cell response specific for tumor-associated antigens other than WT1. Our study provides an approach that targets tumor-specific intracellular antigens without using cell therapy and suggests that epitope spreading could contribute to the therapeutic efficacy of this BiTE.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Proteínas Repressoras/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Humanos , Camundongos , Terapia de Alvo Molecular/métodos , Proteínas Oncogênicas/imunologia , Resultado do Tratamento , Proteínas WT1
18.
Clin Cancer Res ; 20(15): 4036-46, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24850840

RESUMO

PURPOSE: RMFPNAPYL (RMF), a Wilms' tumor gene 1 (WT1)-derived CD8 T-cell epitope presented by HLA-A*02:01, is a validated target for T-cell-based immunotherapy. We previously reported ESK1, a high avidity (Kd < 0.2 nmol/L), fully-human monoclonal antibody (mAb) specific for the WT1 RMF peptide/HLA-A*02:01 complex, which selectively bound and killed WT1(+) and HLA-A*02:01(+) leukemia and solid tumor cell lines. EXPERIMENTAL DESIGN: We engineered a second-generation mAb, ESKM, to have enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) function due to altered Fc glycosylation. ESKM was compared with native ESK1 in binding assays, in vitro ADCC assays, and mesothelioma and leukemia therapeutic models and pharmacokinetic studies in mice. ESKM toxicity was assessed in HLA-A*02:01(+) transgenic mice. RESULTS: ESK antibodies mediated ADCC against hematopoietic and solid tumor cells at concentrations below 1 µg/mL, but ESKM was about 5- to 10-fold more potent in vitro against multiple cancer cell lines. ESKM was more potent in vivo against JMN mesothelioma, and effective against SET2 AML and fresh ALL xenografts. ESKM had a shortened half-life (4.9 days vs. 6.5 days), but an identical biodistribution pattern in C57BL/6J mice. At therapeutic doses of ESKM, there was no difference in half-life or biodistribution in HLA-A*02:01(+) transgenic mice compared with the parent strain. Importantly, therapeutic doses of ESKM in these mice caused no depletion of total WBCs or hematopoetic stem cells, or pathologic tissue damage. CONCLUSIONS: The data provide proof of concept that an Fc-enhanced mAb can improve efficacy against a low-density, tumor-specific, peptide/MHC target, and support further development of this mAb against an important intracellular oncogenic protein.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno HLA-A2/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Leucemia Experimental/terapia , Mesotelioma/terapia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Citotóxicos/imunologia , Proteínas WT1/imunologia , Animais , Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Citometria de Fluxo , Humanos , Imunoterapia , Leucemia Experimental/imunologia , Masculino , Mesotelioma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Células Tumorais Cultivadas
19.
Sci Transl Med ; 5(176): 176ra33, 2013 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23486779

RESUMO

The Wilms tumor 1 (WT1) oncoprotein is an intracellular, oncogenic transcription factor that is overexpressed in a wide range of leukemias and solid cancers. RMFPNAPYL (RMF), a WT1-derived CD8+ T cell human leukocyte antigen (HLA)-A0201 epitope, is a validated target for T cell-based immunotherapy. Using phage display technology, we discovered a fully human "T cell receptor-like" monoclonal antibody (mAb), ESK1, specific for the WT1 RMF peptide/HLA-A0201 complex. ESK1 bound to several leukemia and solid tumor cell lines and primary leukemia cells, in a WT1- and HLA-A0201-restricted manner, with high avidity [dissociation constant (Kd)=0.1 nM]. ESK1 mediated antibody-dependent human effector cell cytotoxicity in vitro. Low doses of naked ESK1 antibody cleared established, disseminated, human acute lymphocytic leukemia and Philadelphia chromosome-positive leukemia in nonobese diabetic/severe combined immunodeficient γc-/- (NSG) mouse models. At therapeutic doses, no toxicity was seen in HLA-A0201 transgenic mice. ESK1 is a potential therapeutic agent for a wide range of cancers overexpressing the WT1 oncoprotein. This finding also provides preclinical validation for the strategy of developing therapeutic mAbs targeting intracellular oncogenic proteins.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Tumor de Wilms/terapia , Animais , Linfócitos T CD8-Positivos/metabolismo , Epitopos/imunologia , Humanos , Leucemia/imunologia , Leucemia/terapia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Linfócitos T Citotóxicos/imunologia , Tumor de Wilms/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa