Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Mol Cancer Ther ; 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38647528

RESUMO

Mesothelin (MSLN) is a cell-surface protein that is expressed on many cancers, which makes it a popular target for antibody-based cancer therapy. However, MSLN is shed from cancer cells at high levels via proteases that cleave at its membrane-proximal C-terminal region. Shed MSLN accumulates in patient fluids and tumors and can block antibody-based MSLN-targeting drugs from killing cancer cells. A previously established monoclonal antibody (mAb), 15B6, binds MSLN at its protease-sensitive C-terminal region and does not bind shed MSLN. 15B6 variable fragment (Fv)-derived chimeric antigen receptor (CAR) T cells are not inhibited by shed MSLN and kill tumors in mice more effectively than mAb SS1 Fv-derived CAR T cells, which bind an epitope retained in shed MSLN. Here, we have established 15B6 Fv-derived MSLN x CD3 bispecific antibodies (BsAbs) that target MSLN-expressing cancers. We identified our lead candidate, BsAb 5, after screening multiple 15B6-derived BsAb formats in vitro for cytotoxic activity. BsAb 5 activates T cells to kill various cancer cell lines in a MSLN-specific manner. MSLN 296-591 His, a recombinant protein mimicking shed MSLN, does not inhibit 15B6-derived BsAb 5 but completely inhibits humanized SS1-derived BsAb 7. Furthermore, BsAb 5 inhibits and delays tumor growth and is not inhibited by MSLN 296-585 His in mice. Our findings indicate that by targeting the protease-sensitive region of MSLN, BsAb 5 has high MSLN-specific anticancer activity that is not inhibited by shed MSLN. BsAb 5 may be a promising immunotherapy candidate for MSLN-expressing cancers.

2.
Proc Natl Acad Sci U S A ; 119(19): e2202439119, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35512094

RESUMO

SignificanceMesothelin (MSLN) is a cell-surface protein that is a popular target for antibody-based therapies. We have identified shed MSLN as a major obstacle to successful antibody therapies and prepared a monoclonal antibody that inhibits shedding and makes very active CAR T cells whose activity is not blocked by shed MSLN and merits further preclinical development.


Assuntos
Receptores de Antígenos Quiméricos , Anticorpos Monoclonais/metabolismo , Linhagem Celular Tumoral , Proteínas Ligadas por GPI/metabolismo , Mesotelina , Linfócitos T
3.
Br J Cancer ; 126(5): 754-763, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34876673

RESUMO

BACKGROUND: Thymic epithelial tumours (TETs) are rare tumours comprised of thymomas and thymic carcinoma. Novel therapies are needed, especially in thymic carcinoma where the 5-year survival rate hovers at 30%. Mesothelin (MSLN), a surface glycoprotein that is cleaved to produce mature MSLN (mMSLN) and megakaryocyte potentiating factor (MPF), is expressed in limited tissues. However, its expression is present in various cancers, including thymic carcinoma, where it is expressed in 79% of cases. METHODS: We utilised flow cytometry, in vitro cytotoxicity assays, and an in vivo xenograft model in order to demonstrate the ability of the MSLN targeting antibody-drug conjugate (ADC) anetumab ravtansine (ARav) in inhibiting the growth of thymic carcinoma. RESULTS: Thymoma and thymic carcinoma cell lines express MSLN, and anetumab, the antibody moiety of ARav, was capable of binding MSLN expressing thymic carcinoma cells and internalising. ARav was effective at inhibiting the growth of thymic carcinoma cells stably transfected with mMSLN in vitro. In vivo, 15 mg/kg ARav inhibited T1889 xenograft tumour growth, while combining 7.5 mg/kg ARav with 4 mg/kg cisplatin yielded an additive effect on inhibiting tumour growth. CONCLUSIONS: These data demonstrate that anetumab ravtansine inhibits the growth of MSLN positive thymic carcinoma cells in vitro and in vivo.


Assuntos
Imunoconjugados/administração & dosagem , Maitansina/análogos & derivados , Mesotelina/genética , Mesotelina/metabolismo , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Timoma/tratamento farmacológico , Neoplasias do Timo/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Imunoconjugados/farmacologia , Maitansina/administração & dosagem , Maitansina/farmacologia , Camundongos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Timoma/genética , Timoma/metabolismo , Neoplasias do Timo/genética , Neoplasias do Timo/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Proc Natl Acad Sci U S A ; 116(10): 4575-4582, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30760587

RESUMO

The tumor microenvironment plays a critical role in controlling tumor progression and immune surveillance. We produced an immunotoxin (2E4-PE38) that kills mouse cells expressing CD25 by attaching the Fv portion of monoclonal antibody 2E4 (anti-mouse CD25) to a 38-kDa portion of Pseudomonas exotoxin A. We employed three mouse cancer tumor models (AB1 mesothelioma, 66c14 breast cancer, and CT26M colon cancer). Tumors were implanted at two sites on BALB/c mice. On days 5 and 9, one tumor was directly injected with 2E4-PE38, and the other was not treated; 2E4-PE38 produced complete regressions of 85% of injected AB1 tumors, 100% of 66c14 tumors, and 100% of CT26M tumors. It also produced complete regressions of 77% of uninjected AB1 tumors, 47% of 66c14 tumors, and 92% of CT26M tumors. Mice with complete regressions of 66c14 tumors were immune to rechallenge with 66c14 cells. Mice with complete regressions of AB1 or CT26M tumors developed cross-tumor immunity rejecting both tumor types. Injection of anti-CD25 antibody or a mutant inactive immunotoxin were generally ineffective. Tumors were analyzed 3 days after 2E4-PE38 injection. The number of regulatory T cells (Tregs) was significantly reduced in the injected tumor but not in the spleen. Injected tumors contained an increase in CD8 T cells expressing IFN-γ, the activation markers CD69 and CD25, and macrophages and conventional dendritic cells. Treatment with antibodies to CD8 abolished the antitumor effect. Selective depletion of Tregs in tumors facilitates the development of a CD8 T cell-dependent antitumor effect in three mouse models.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Imunoterapia , Imunotoxinas/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , ADP Ribose Transferases/imunologia , Animais , Toxinas Bacterianas/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Exotoxinas/imunologia , Feminino , Humanos , Imunidade/efeitos dos fármacos , Interferon gama/imunologia , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/citologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Fatores de Virulência/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
5.
JCO Precis Oncol ; 20182018.
Artigo em Inglês | MEDLINE | ID: mdl-30370397

RESUMO

PURPOSE: Effective biomarkers for malignant mesothelioma (MM) are needed for clinical management and the development of mesothelin-targeted therapies. We evaluated serum megakaryocyte potentiating factor (MPF) as a biomarker predictive of treatment outcome in patients with MM and for developing mesothelin-targeted therapies. MATERIALS AND METHODS: Serial serum samples from patients with MM in two clinical trials of an antimesothelin immunotoxin were tested with our clinically validated MPF assay. Correlative studies were performed to determine the test effectiveness in treatment monitoring and outcome prediction. MPF was further evaluated for an association with response to an antimesothelin therapy and for disease monitoring. RESULTS: There was a significant reduction of serum MPF in patients with elevated baseline and radiologic response, with an average change from -52% to -78% after one to six cycles. Using a -50% change as the cutoff, patients with MM with positive MPF response had significantly improved progression-free survival (P < .001), with the median extended from 1.9 to 11.3 months. These patients with MPF response further exhibited improved overall survival (P = .004), with the median extended from 8.8 to 22.3 months. In patients with refractory MM, there was an association between elevated pretreatment serum MPF and radiologic response to an antimesothelin therapy (P = .033). Furthermore, in these response patients, serum MPF was monitored between 32.2 and 63.8 months and was found to reflect treatment response and disease progression. CONCLUSION: At a cutoff of -50% change after receiving systemic therapies, a reduction in MPF was associated with improved clinical outcome, both progression-free survival and overall survival. An elevated baseline serum MPF was associated with a response to an antimesothelin therapy in patients with refractory MM; however, this finding needs to be confirmed in another study.

6.
J Appl Lab Med ; 3(2): 166-177, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30370398

RESUMO

Background: There is an urgent need for a companion assay to work with mesothelin-targeted therapeutic agents and for noninvasive and accurate prognostication of malignant mesothelioma (MM) patients. We report the development and validation of a blood-based assay for megakaryocyte potentiating factor (MPF) and the evaluation of its effectiveness for prognosis in MM and lung cancer patients. Methods: Using electrochemiluminescence technology, we developed a sensitive MPF assay and performed both analytical and clinical validations. Further, the effectiveness of the MPF assay in predicting prognosis was evaluated for 95 MM and 272 lung cancer patients. Results: We performed comprehensive analytical and clinical validation, including precision and accuracy, interference, preanalytical variables, sensitivity, and specificity for mesothelioma. In MM patients, increased serum MPF is a predictor of poor survival with a hazard ratio (HR) = 2.46 (log-rank P = 0.003; n = 95). In refractory MM patients, increased MPF is a strong predictor of poor outcome with an HR = 6.12 (log-rank P = 0.0007; n = 57). In a lung cancer patient cohort, increased MPF is a predictor of poor survival, with an HR = 1.57 (log-rank P = 0.003; n = 272). Conclusions: The MPF assay has robust technical characteristics, with strong analytic and clinical validation. Clinical studies indicate that increased serum MPF is a predictor of poor survival for MM patients, throughout the course of the disease. Increased MPF is also associated with poor overall survival for patients with newly diagnosed lung cancer.


Assuntos
Proteínas Ligadas por GPI/sangue , Neoplasias Pulmonares , Mesotelioma , Biomarcadores Tumorais/sangue , Estudos de Coortes , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/diagnóstico , Mesotelina , Mesotelioma/sangue , Mesotelioma/diagnóstico , Mesotelioma Maligno , Prognóstico , Sensibilidade e Especificidade , Análise de Sobrevida
7.
Proc Natl Acad Sci U S A ; 115(15): E3501-E3508, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581296

RESUMO

Recombinant immunotoxins (RITs) are chimeric proteins consisting of a Fv that binds to a cancer cell and a portion of a protein toxin. One of these, Moxetumomab pasudotox, was shown to be effective in treating patients with some leukemias, where the cells are readily accessible to the RIT. However, their short half-life limits their efficacy in solid tumors, because penetration into the tumors is slow. Albumin and agents bound to albumin have a long half-life in the circulation. To increase the time tumor cells are exposed to RITs, we have produced and evaluated variants that contain either an albumin-binding domain (ABD) from Streptococcus or single-domain antibodies from Llama. We have inserted these ABDs into RITs targeting mesothelin, between the Fv and the furin cleavage site. We find that these proteins can be produced in large amounts, are very cytotoxic to mesothelin-expressing cancer cell lines, and have a high affinity for human or mouse serum albumin. In mice, the RIT containing an ABD from Streptococcus has a longer half-life and higher antitumor activity than the other two. Its half-life in the circulation of mice ranges from 113 to 194 min compared with 13 min for an RIT with no ABD. Cell uptake studies show the RIT enters the target cell bound to serum albumin. We conclude that RITs with improved half-lives and antitumor activity should be evaluated for the treatment of cancer in humans.


Assuntos
Imunotoxinas/farmacocinética , Animais , Toxinas Bacterianas/farmacocinética , Toxinas Bacterianas/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Modelos Animais de Doenças , Exotoxinas/farmacocinética , Exotoxinas/farmacologia , Feminino , Proteínas Ligadas por GPI/efeitos dos fármacos , Meia-Vida , Humanos , Imunotoxinas/imunologia , Leucemia/tratamento farmacológico , Mesotelina , Camundongos , Camundongos Nus , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Albumina Sérica/metabolismo , Albumina Sérica/uso terapêutico
8.
Proc Natl Acad Sci U S A ; 115(4): E733-E742, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29311317

RESUMO

Protein-based drugs are very active in treating cancer, but their efficacy can be limited by the formation of neutralizing antidrug antibodies (ADAs). Recombinant immunotoxins are proteins that are very effective in patients with leukemia, where immunity is suppressed, but induce ADAs, which compromise their activity, in patients with intact immunity. Here we induced a specific, durable, and transferable immune tolerance to recombinant immunotoxins by combining them with nanoparticles containing rapamycin (SVP-R). SVP-R mitigated the formation of inhibitory ADAs in naïve and sensitized mice, resulting in restoration of antitumor activity. The immune tolerance is mediated by colocalization of the SVP-R and immunotoxin to dendritic cells and macrophages in the spleen and is abrogated by depletion of regulatory T cells. Tolerance induced by SVPs was not blocked by checkpoint inhibitors or costimulatory agonist monoclonal antibodies that by themselves enhance ADA formation.


Assuntos
Imunomodulação , Imunossupressores/administração & dosagem , Imunotoxinas/administração & dosagem , Leucemia/terapia , Sirolimo/administração & dosagem , Animais , Anticorpos Neutralizantes , Proteínas Ligadas por GPI/imunologia , Humanos , Imunotoxinas/imunologia , Mesotelina , Nanopartículas , Fatores de Tempo
9.
Oncotarget ; 8(16): 26744-26754, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28460459

RESUMO

Mesothelin (MSLN) is a glycophosphatidylinositol (GPI)-linked cell surface protein highly expressed in several types of malignant tumors sometimes in association with increased tumor aggressiveness and poor clinical outcome. In the present study, 1562 tumors were immunohistochemically analyzed for mesothelin expression using two different types of mouse monoclonal antibodies (5B2 and MN-1) to determine the clinical usefulness of mesothelin immunohistochemistry as well as to pinpoint potential targets for future anti-mesothelin therapy. Also, characterization of selected mesothelin-positive tumors was performed by immunohistochemistry and oncogene sequencing. Among the tumors analyzed, the highest frequencies of mesothelin-positivity were detected in ovarian serous carcinoma (90% in 5B2 and 94% in MN-1). Both antibodies showed frequent positivity in pancreatic adenocarcinoma (71% using 5B2 and 87% using MN-1) and malignant pleural mesothelioma (75% using 5B2 and 78% using MN-1). In malignant mesothelioma, overall survival was significantly longer in the cohort of patients with diffuse membranous expression of mesothelin (P < 0.001). Both antibodies showed positive staining in thymic carcinoma (77% in 5B2 and 59% in MN-1), however, no expression was detected in thymoma. No correlation was detected between mesothelin expression and mismatch repair system deficient phenotype or gene mutation (BRAF and RAS) status in gastrointestinal adenocarcinomas. Mesothelin immunohistochemistry may assist the differential diagnosis of thymoma vs. thymic carcinoma as well as prognostication of mesothelioma patients. Our results demonstrate that patients with solid tumors expressing mesothelin could be targeted by anti-mesothelin therapies.


Assuntos
Biomarcadores Tumorais , Proteínas Ligadas por GPI/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Mesotelioma/metabolismo , Mesotelioma/mortalidade , Neoplasias Pleurais/metabolismo , Neoplasias Pleurais/mortalidade , Anticorpos Monoclonais , Diagnóstico Diferencial , Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Mesotelina , Mesotelioma/diagnóstico , Mesotelioma/genética , Mesotelioma Maligno , Mutação , Neoplasias Pleurais/diagnóstico , Neoplasias Pleurais/genética , Prognóstico , Análise de Sobrevida
10.
Mol Oncol ; 10(8): 1317-29, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27507537

RESUMO

Mesothelin overexpression in lung adenocarcinomas correlates with the presence of activating KRAS mutations and poor prognosis. Hence SS1P, a mesothelin-targeted immunotoxin, could offer valuable treatment options for these patients, but its use in solid tumor therapy is hampered by high immunogenicity and non-specific toxicity. To overcome both obstacles we developed RG7787, a de-immunized cytotoxic fusion protein comprising a humanized SS1 Fab fragment and a truncated, B-cell epitope silenced, 24 kD fragment of Pseudomonas exotoxin A (PE24). Reactivity of RG7787 with sera from immunotoxin-treated patients was >1000 fold reduced. In vitro RG7787 inhibited cell viability of lung cancer cell lines with picomolar potency. The pharmacokinetic properties of RG7787 in rodents were comparable to SS1P, yet it was tolerated up to 10 fold better without causing severe vascular leak syndrome or hepatotoxicity. A pharmacokinetic/pharmacodynamic model developed based on NCI-H596 xenograft studies showed that for RG7787 and SS1P, their in vitro and in vivo potencies closely correlate. At optimal doses of 2-3 mg/kg RG7787 is more efficacious than SS1P. Even large, well established tumors (600 mm(3)) underwent remission during three treatment cycles with RG7787. Also in two patient-derived lung cancer xenograft models, Lu7336 and Lu7187, RG7787 showed anti-tumor efficacy. In monotherapy two treatment cycles were moderately efficacious in the Lu7336 model but showed good anti-tumor activity in the KRAS mutant Lu7187 model (26% and 80% tumor growth inhibition, respectively). Combination of RG7787 with standard chemotherapies further enhanced efficacy in both models achieving near complete eradication of Lu7187 tumors.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Proteínas Ligadas por GPI/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Engenharia de Proteínas , Pseudomonas/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Fatores de Virulência/uso terapêutico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Pulmonares/patologia , Mesotelina , Camundongos SCID , Modelos Biológicos , Ratos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
11.
Toxins (Basel) ; 8(8)2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27463727

RESUMO

Recombinant immunotoxins (RITs) are fusions of an Fv-based targeting moiety and a toxin. Pseudomonas exotoxin A (PE) has been used to make several immunotoxins that have been evaluated in clinical trials. Immunogenicity of the bacterial toxin and off-target toxicity have limited the efficacy of these immunotoxins. To address these issues, we have previously made RITs in which the Fv is connected to domain III (PE24) by a furin cleavage site (FCS), thereby removing unneeded sequences of domain II. However, the PE24 containing RITs do not contain the naturally occurring disulfide bond around the furin cleavage sequence, because it was removed when domain II was deleted. This could potentially allow PE24 containing immunotoxins to be cleaved and inactivated before internalization by cell surface furin or other proteases in the blood stream or tumor microenvironment. Here, we describe five new RITs in which a disulfide bond is engineered to protect the FCS. The most active of these, SS1-Fab-DS3-PE24, shows a longer serum half-life than an RIT without the disulfide bond and has the same anti-tumor activity, despite being less cytotoxic in vitro. These results have significance for the production of de-immunized, low toxicity, PE24-based immunotoxins with a longer serum half-life.


Assuntos
ADP Ribose Transferases/farmacologia , Toxinas Bacterianas/farmacologia , Proliferação de Células/efeitos dos fármacos , Dissulfetos/farmacologia , Desenho de Fármacos , Exotoxinas/farmacologia , Furina/metabolismo , Região Variável de Imunoglobulina/farmacologia , Imunotoxinas/farmacologia , Neoplasias/tratamento farmacológico , Fatores de Virulência/farmacologia , ADP Ribose Transferases/sangue , ADP Ribose Transferases/química , Animais , Toxinas Bacterianas/sangue , Toxinas Bacterianas/química , Linhagem Celular Tumoral , Dissulfetos/sangue , Dissulfetos/química , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Exotoxinas/sangue , Exotoxinas/química , Meia-Vida , Humanos , Região Variável de Imunoglobulina/sangue , Região Variável de Imunoglobulina/química , Imunotoxinas/sangue , Imunotoxinas/química , Concentração Inibidora 50 , Mesotelina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Oxirredução , Domínios Proteicos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Relação Estrutura-Atividade , Microambiente Tumoral , Fatores de Virulência/sangue , Fatores de Virulência/química , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
12.
Oncotarget ; 7(21): 29916-26, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27167198

RESUMO

Recombinant immunotoxins (RITs) are genetically engineered proteins being developed to treat cancer. They are composed of an Fv that targets a cancer antigen and a portion of a protein toxin. Their clinical success is limited by their immunogenicity. Our goal is to produce a new RIT that targets mesothelin and is non-immunogenic by combining mutations that decrease B- and T-cell epitopes. Starting with an immunotoxin that has B-cell epitopes suppressed, we added mutations step-wise that suppress T-cell epitopes. The final protein (LMB-T14) has greatly reduced antigenicity as assessed by binding to human anti-sera and a greatly decreased ability to activate helper T-cells evaluated in a T-cell activation assay. It is very cytotoxic to mesothelioma cells from patients, and to cancer cell lines. LMB-T14 produces complete remissions of a mesothelin expressing cancer (A431/H9) xenograft. The approach used here can be used to de-immunize other therapeutic foreign proteins.


Assuntos
Proteínas Ligadas por GPI/antagonistas & inibidores , Imunotoxinas/imunologia , Neoplasias Pulmonares/terapia , Mesotelioma/terapia , Proteínas Recombinantes de Fusão/imunologia , Imunidade Adaptativa , Animais , Anticorpos Monoclonais/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Linhagem Celular Tumoral , Epitopos de Linfócito B/genética , Epitopos de Linfócito T/genética , Exotoxinas/genética , Exotoxinas/imunologia , Feminino , Humanos , Região Variável de Imunoglobulina/imunologia , Imunoterapia/métodos , Imunotoxinas/genética , Imunotoxinas/uso terapêutico , Neoplasias Pulmonares/imunologia , Mesotelina , Mesotelioma/imunologia , Mesotelioma Maligno , Camundongos , Camundongos Nus , Terapia de Alvo Molecular/métodos , Mutação , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Auxiliares-Indutores , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Immunol Rev ; 270(1): 152-64, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26864110

RESUMO

Recombinant immunotoxins (RITs) are chimeric proteins designed to treat cancer. They are made up of an Fv or Fab that targets an antigen on a cancer cell fused to a 38-kDa portion of Pseudomonas exotoxin A (PE38). Because PE38 is a bacterial protein, it is highly immunogenic in patients with solid tumors that have normal immune systems, but much less immunogenic in patients with hematologic malignancies where the immune system is suppressed. RITs have shown efficacy in refractory hairy cell leukemia and in some children with acute lymphoblastic leukemia, but have been much less effective in solid tumors, because neutralizing antibodies develop and prevent additional treatment cycles. In this paper we will (i) review data from clinical trials describing the immunogenicity of PE38 in different patient populations; (ii) review results from clinical trials using different immunosuppressive drugs; and (iii) describe our efforts to make new less-immunogenic RITs by identifying and removing T- and B-cell epitopes to hide the RIT from the immune system.


Assuntos
Imunotoxinas/imunologia , Imunotoxinas/uso terapêutico , Proteínas Recombinantes de Fusão , ADP Ribose Transferases/química , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Neutralizantes/imunologia , Formação de Anticorpos , Antígenos/imunologia , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Ensaios Clínicos como Assunto , Vias de Administração de Medicamentos , Quimioterapia Combinada , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Exotoxinas/química , Exotoxinas/genética , Exotoxinas/imunologia , Engenharia Genética , Humanos , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/uso terapêutico , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Imunoterapia , Imunotoxinas/administração & dosagem , Imunotoxinas/efeitos adversos , Imunotoxinas/química , Imunotoxinas/genética , Mesotelina , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Polietilenoglicóis , Deleção de Sequência , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
14.
J Immunol ; 194(4): 1695-701, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25560410

RESUMO

Recombinant immunotoxin (RIT) therapy is limited in patients by neutralizing Ab responses. Ninety percent of patients with normal immune systems make neutralizing Abs after one cycle of RIT, preventing repeated dosing. Furthermore, some patients have pre-existing Abs from environmental exposure to Pseudomonas exotoxin, the component of the RIT that elicits the neutralizing Ab response. Bortezomib is an U.S. Food and Drug Administration-approved proteasome inhibitor that selectively targets and kills plasma cells that are necessary for the neutralizing Ab response. We hypothesized that bortezomib may abrogate neutralizing Ab levels, making dosing of RIT possible in mice already immune to RIT. We immunized BALB/c mice with multiple doses of SS1P, a RIT whose Ab portion targets mesothelin. Mice with elevated Ab levels were separated into groups to receive saline, bortezomib, the pentostatin/cyclophosphamide (PC) regimen, or the bortezomib/PC (BPC) combination regimen. Four weeks after finishing therapy, plasma Ab levels were assayed, and bone marrow was harvested. The bortezomib and PC regimens significantly reduced Ab levels, and we observed fewer plasma cells in the bone marrow of bortezomib-treated mice but not in PC-treated mice. The BPC combination regimen almost completely eliminated Abs and further reduced plasma cells in the bone marrow. This regimen is more effective than individual regimens and may reduce Ab levels in patients with pre-existing neutralizing Abs to Pseudomonas exotoxin, allowing RIT treatment.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/sangue , Ácidos Borônicos/farmacologia , Inibidores de Proteassoma/farmacologia , Pirazinas/farmacologia , ADP Ribose Transferases/imunologia , Animais , Toxinas Bacterianas/imunologia , Bortezomib , Ensaio de Imunoadsorção Enzimática , Exotoxinas/imunologia , Feminino , Imunoterapia/métodos , Mesotelina , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/imunologia , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
15.
Leuk Res ; 38(10): 1224-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25127689

RESUMO

Moxetumomab pasudotox (HA22) is a recombinant immunotoxin, now in clinical trials, that combines an anti-CD22-Fv with a 38-kDa fragment of Pseudomonas exotoxin A. To produce a less immunogenic molecule without reducing the half-life in circulation, we constructed LMB11 combining an anti-CD22 Fab with a less immunogenic version of PE38. We found that LMB11 retains full activity toward CD22-expressing cells. In mice, the half-life of LMB11 is 29 min and the antitumor activity of LMB11 is better than that of HA22. Because it can be safely given at much higher doses, LMB11 produced complete tumor remissions in 7/7 mice.


Assuntos
Fragmentos Fab das Imunoglobulinas/farmacologia , Imunotoxinas/farmacologia , Leucemia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Toxinas Bacterianas , Exotoxinas , Feminino , Meia-Vida , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Immunol ; 193(1): 48-55, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24890727

RESUMO

Immunogenicity remains the "Achilles' heel" of protein-based therapeutics. Anti-drug Abs produced in response to protein therapeutics can severely limit both the safety and efficacy of this expanding class of agent. In this article, we report that monotherapy of mice with tofacitinib (the JAK inhibitor) quells Ab responses to an immunotoxin derived from the bacterial protein Pseudomonas exotoxin A, as well as to the model Ag keyhole limpet hemocyanin. Thousand-fold reductions in IgG1 titers to both Ags were observed 21 d post immunization. In fact, suppression was evident for all IgG isotypes and IgM. A reduction in IgG3 production was also noted with a thymus-independent type II Ag. Mechanistic investigations revealed that tofacitinib treatment led to reduced numbers of CD127+ pro-B cells. Furthermore, we observed fewer germinal center B cells and the impaired formation of germinal centers of mice treated with tofacitinib. Because normal Ig levels were still present during tofacitinib treatment, this agent specifically reduced anti-drug Abs, thus preserving the potential efficacy of biological therapeutics, including those used as cancer therapeutics.


Assuntos
ADP Ribose Transferases/farmacologia , Formação de Anticorpos/efeitos dos fármacos , Toxinas Bacterianas/farmacologia , Exotoxinas/farmacologia , Hemocianinas/farmacologia , Imunotoxinas/farmacologia , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Fatores de Virulência/farmacologia , Animais , Linfócitos B/imunologia , Feminino , Centro Germinativo/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Exotoxina A de Pseudomonas aeruginosa
17.
Proc Natl Acad Sci U S A ; 111(23): 8571-6, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24799704

RESUMO

Nonhuman proteins have valuable therapeutic properties, but their efficacy is limited by neutralizing antibodies. Recombinant immunotoxins (RITs) are potent anticancer agents that have produced many complete remissions in leukemia, but immunogenicity limits the number of doses that can be given to patients with normal immune systems. Using human cells, we identified eight helper T-cell epitopes in PE38, a portion of the bacterial protein Pseudomonas exotoxin A which consists of the toxin moiety of the RIT, and used this information to make LMB-T18 in which three epitopes were deleted and five others diminished by point mutations in key residues. LMB-T18 has high cytotoxic and antitumor activity and is very resistant to thermal denaturation. The new immunotoxin has a 93% decrease in T-cell epitopes and should have improved efficacy in patients because more treatment cycles can be given. Furthermore, the deimmunized toxin can be used to make RITs targeting other antigens, and the approach we describe can be used to deimmunize other therapeutically useful nonhuman proteins.


Assuntos
Epitopos de Linfócito T/imunologia , Imunotoxinas/imunologia , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/imunologia , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Aminoácidos/genética , Aminoácidos/imunologia , Animais , Formação de Anticorpos/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Eletroforese em Gel de Poliacrilamida , Mapeamento de Epitopos , Exotoxinas/genética , Exotoxinas/imunologia , Feminino , Humanos , Imunoterapia/métodos , Imunotoxinas/genética , Imunotoxinas/uso terapêutico , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos SCID , Modelos Moleculares , Neoplasias/patologia , Neoplasias/terapia , Peptídeos/genética , Peptídeos/imunologia , Mutação Puntual , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
18.
Mol Cancer Ther ; 12(1): 48-57, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23136186

RESUMO

SS1P is a recombinant immunotoxin (RIT) engineered for the targeted elimination of malignant cells that express the tumor-associated antigen mesothelin. It is composed of an antimesothelin antibody variable fragment (Fv) linked to a cytotoxic fragment of Pseudomonas exotoxin A (PE) that includes domains II and III of native PE. The clinical use of SS1P is limited by its propensity to induce neutralizing antibodies and to cause a dose-limiting capillary leak syndrome (CLS) in patients. In this article, we describe a reengineered SS1P with improved properties that overcome these deficits. The redesign of SS1P consists of (i) removing the bulk of PE domain II (residues 251-273 and 284-394 of native PE), leaving only an 11-residue furin cleavage site, (ii) adding a Gly-Gly-Ser peptide linker after the furin cleavage site, and (iii) replacing eight highly solvent-exposed residues in the catalytic domain of PE. The new molecule, SS1-LR/GGS/8M, has cytotoxic activity comparable with SS1P on several mesothelin-expressing cell lines and remarkably improved activity on primary cells from patients with mesothelioma. In a mouse xenograft tumor model, high doses of SS1-LR/GGS/8M elicit antitumor activity superior to the activity of SS1P at its maximum-tolerated dose. In addition, SS1-LR/GGS/8M has greatly decreased ability to cause CLS in a rat model and reduced antigenicity or reactivity with antibodies to the sera of patients previously treated with SS1P.


Assuntos
Antineoplásicos/farmacologia , Proteínas Ligadas por GPI/metabolismo , Mesotelioma/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Substituição de Aminoácidos , Animais , Antígenos de Neoplasias , Antineoplásicos/efeitos adversos , Antineoplásicos/imunologia , Síndrome de Vazamento Capilar/induzido quimicamente , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI/imunologia , Meia-Vida , Humanos , Concentração Inibidora 50 , Dose Máxima Tolerável , Mesotelina , Mesotelioma/patologia , Camundongos , Camundongos Nus , Mutagênese Sítio-Dirigida , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Methods Mol Biol ; 907: 627-43, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22907377

RESUMO

Recombinant immunotoxin (RIT) contains the Fv portion of the antibody fused to the truncated form of toxin and are ongoing in clinical trials for cancer therapy. To obtain high yields of products, RITs are produced in Escherichia coli (E. coli). As the endotoxin came from E. coli cells and is harmful to animals, it is important to produce the RITs with low endotoxin. This section describes the protocols to produce RITs containing low level of endotoxins.


Assuntos
Endotoxinas/biossíntese , Imunotoxinas/metabolismo , Biologia Molecular/métodos , Proteínas Recombinantes/biossíntese , Animais , Antígenos/imunologia , Bioensaio , Morte Celular , Linhagem Celular , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Fermentação , Humanos , Corpos de Inclusão/metabolismo , Lipopolissacarídeos/metabolismo , Plasmídeos/metabolismo , Desnaturação Proteica , Redobramento de Proteína , Solubilidade , Transformação Genética
20.
Proc Natl Acad Sci U S A ; 109(29): 11782-7, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22753489

RESUMO

Recombinant immunotoxins (RITs) are hybrid proteins used to treat cancer. These proteins are composed of an Fv that reacts with cancer cells joined to a portion of Pseudomonas exotoxin A, which kills the cell. Because the toxin is a foreign protein, it can induce neutralizing antibodies and thereby limit the number of doses a patient can receive. We previously identified seven major mouse B-cell epitopes in the toxin, and subsequently silenced them using point mutations that converted large hydrophilic amino acids to alanine, yet retained full antitumor activity. Here we present results in which we identify and silence human B-cell epitopes in the RIT HA22. We obtained B cells from patients with antibodies to RITs, isolated the corresponding variable fragments (Fvs), and constructed a phage-display library containing Fvs that bind to the RITs. We then used alanine scanning mutagenesis to locate the epitopes. We found that human and mouse epitopes frequently overlap but are not identical. Most mutations that remove mouse epitopes did not remove human epitopes. Using the epitope information, we constructed a variant immunotoxin, HA22-LR-LO10, which has low reactivity with human antisera, yet has high cytotoxic and antitumor activity and can be given to mice at high doses without excess toxicity. The toxin portion of this RIT (LR-LO10) can be used with Fvs targeting other cancer antigens and is suitable for clinical development.


Assuntos
Antineoplásicos/imunologia , Epitopos de Linfócito B/imunologia , Imunoterapia/métodos , Imunotoxinas/imunologia , Modelos Moleculares , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/imunologia , Sequência de Aminoácidos , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Imunotoxinas/química , Imunotoxinas/genética , Imunotoxinas/farmacocinética , Camundongos , Dados de Sequência Molecular , Mutagênese , Neoplasias/imunologia , Biblioteca de Peptídeos , Engenharia de Proteínas/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Anticorpos de Cadeia Única/imunologia , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA