Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Anal Biochem ; 653: 114776, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35679954

RESUMO

Targeted tumor therapy is an attractive approach for cancer treatment. Delta-like ligand 4 (DLL4) is overexpressed in tumor vasculature and plays a pivotal role in tumor neovascular development and angiogenesis during tumor progression. Immunotoxins due to their superior cell-killing ability and the relative simplicity of their preparation, have great potential in the clinical treatment of cancer. The aim of this study was to develop a novel immunotoxin against DLL4 as a cell cytotoxic agent and angiogenesis maturation inhibitor. In present study, an immunotoxin, named DLL4Nb-PE, in which a Nanobody as targeting moiety fused to the Pseudomonas exotoxin A (PE) was constructed, expressed and assessed by SDS-PAGE, western blotting, ELISA and flowcytometry. The functional assessment was carried out via MTT, apoptosis and chicken chorioallantoic membrane (CAM) assays. It was demonstrated DLL4Nb-PE specifically binds to DLL4 and recognizes DLL4-expressing MKN cells. The cytotoxicity assays showed that this molecule could induce apoptosis and kill DLL4 positive MKN cells. In addition, it inhibited neovascularization in the chicken chorioallantoic membrane. Our findings indicate designed anti-DLL4 immunotoxin has valuable potential for application to the treatment of tumors with high DLL4 expression.


Assuntos
Imunotoxinas , Neoplasias , Proliferação de Células , Exotoxinas/metabolismo , Exotoxinas/farmacologia , Exotoxinas/uso terapêutico , Humanos , Imunotoxinas/farmacologia , Imunotoxinas/uso terapêutico , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Pseudomonas/metabolismo
2.
Hepatology ; 71(5): 1696-1711, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31520528

RESUMO

BACKGROUND AND AIMS: Treatment of hepatocellular carcinomas using our glypican-3 (GPC3)-targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down-regulating the Wnt signaling pathway. However, immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. APPROACH AND RESULTS: To address these concerns, we engineered HN3-based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3-T20, which was modified to remove T-cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B-cell deimmunized immunotoxin (HN3-mPE24) and our original HN3-immunotoxin with a wild-type PE domain (HN3-PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3-T20 having a KD value of 7.4 nM. HN3-T20 retained 73% enzymatic activity when compared with the wild-type immunotoxin in an adenosine diphosphate-ribosylation assay. Interestingly, a real-time cell growth inhibition assay demonstrated that a single dose of HN3-T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10-day experiment. To enhance HN3-T20's serum retention, we tested the effect of adding a streptococcal albumin-binding domain (ABD) and a llama single-domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme-linked immunosorbent assay and found that HN3-ABD-T20 had a 45-fold higher serum half-life than HN3-T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3-ABD-T20-mediated tumor regression at 1 mg/kg. CONCLUSION: These data indicate that ABD-containing deimmunized HN3-T20 immunotoxins are high-potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Carcinoma Hepatocelular/terapia , Exotoxinas/uso terapêutico , Glipicanas/antagonistas & inibidores , Imunotoxinas/uso terapêutico , Neoplasias Hepáticas/terapia , Anticorpos de Domínio Único/uso terapêutico , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/química , ADP Ribose Transferases/farmacologia , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacologia , Linhagem Celular Tumoral , Exotoxinas/química , Exotoxinas/farmacologia , Humanos , Imunotoxinas/química , Imunotoxinas/farmacologia , Camundongos , Camundongos Nus , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia , Fatores de Virulência/química , Fatores de Virulência/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
3.
Mol Pharm ; 18(6): 2285-2297, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33998814

RESUMO

Cholecystokinin-2 receptor (CCK2R) has been proven to be a specific biomarker for colorectal malignancies. Immunotoxins are a valuable class of immunotherapy agents consisting of a targeting element and a bacterial or plant toxin. Previous work demonstrated that targeting CCK2R is a good therapeutic strategy for the treatment of colorectal cancer (CRC). In the present study, we developed a new version of CCK2R-targeting immunotoxin GD9P using a targeted peptide, GD9, as the binding motif and a truncated Pseudomonas exotoxin A (PE38) as the cytokiller. BALB/c nude mice were treated with different doses of GD9P, and pharmacodynamics, pharmacokinetic, and toxicological data were obtained throughout this study. Compared to the parental immunotoxin rCCK8PE38, GD9P exhibited about 1.5-fold yield, higher fluorescence intensity, and increased antitumor activity against human CRC in vitro and in vivo. The IC50 values of GD9P in vitro ranged from 1.61 to 4.55 nM. Pharmacokinetic studies were conducted in mice with a T1/2 of 69.315 min. When tumor-bearing nude mice were treated with GD9P at doses ≥2 mg/kg for five doses, a rapid shrinkage in tumor volume and, in some cases, complete remission was observed. A preliminary safety evaluation demonstrated a good safety profile of GD9P as a Pseudomonas exotoxin A-based immunotherapy. The therapy in combination with oxaliplatin can increase the antitumor efficacy and reduce the toxic side effects caused by chemotherapy. In conclusion, the data support the use of GD9P as a promising immunotherapy targeting CCK2R-expressing colorectal malignancies.


Assuntos
ADP Ribose Transferases/farmacologia , Antineoplásicos/farmacologia , Toxinas Bacterianas/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Exotoxinas/farmacologia , Receptor de Colecistocinina B/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Fatores de Virulência/farmacologia , ADP Ribose Transferases/genética , ADP Ribose Transferases/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/genética , Toxinas Bacterianas/uso terapêutico , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Exotoxinas/genética , Exotoxinas/uso terapêutico , Humanos , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Distribuição Tecidual , Testes de Toxicidade Aguda , Fatores de Virulência/genética , Fatores de Virulência/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
4.
Proc Natl Acad Sci U S A ; 115(8): E1867-E1875, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29432154

RESUMO

Moxetumomab pasudotox (Moxe) is a chimeric protein composed of an anti-CD22 Fv fused to a portion of Pseudomonas exotoxin A and kills CD22-expressing leukemia cells. It is very active in hairy-cell leukemia, but many children with relapsed/refractory acute lymphoblastic leukemia (ALL) either respond transiently or are initially resistant. Resistance to Moxe in cultured cells is due to low expression of diphthamide genes (DPH), but only two of six ALL blast samples from resistant patients had low DPH expression. To develop a more clinically relevant model of resistance, we treated NSG mice bearing KOPN-8 or Reh cells with Moxe. More than 99.9% of the cancer cells were killed by Moxe, but relapse occurred from discrete bone marrow sites. The resistant cells would no longer grow in cell culture and showed major chromosomal changes and changes in phenotype with greatly decreased CD22. RNA deep sequencing of resistant KOPN-8 blasts revealed global changes in gene expression, indicating dedifferentiation toward less-mature B cell precursors, and showed an up-regulation of myeloid genes. When Moxe was combined with 5-azacytidine, resistance was prevented and survival increased to over 5 months in the KOPN-8 model and greatly improved in the Reh model. We conclude that Moxe resistance in mice is due to a new mechanism that could not be observed using cultured cells and is prevented by treatment with 5-azacytidine.


Assuntos
Azacitidina/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Azacitidina/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Medula Óssea , Linhagem Celular Tumoral , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Exotoxinas/administração & dosagem , Humanos , Leucemia , Camundongos , Neoplasias Experimentais , Leucemia-Linfoma Linfoblástico de Células Precursoras , Recidiva
5.
Cancer Sci ; 111(12): 4548-4557, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33058342

RESUMO

Drug resistance represents an obstacle in colorectal cancer (CRC) treatment because of its association with poor prognosis. rBC2LCN is a lectin isolated from Burkholderia that binds cell surface glycans that have fucose moieties. Because fucosylation is enhanced in many types of cancers, this lectin could be an efficient drug carrier if CRC cells specifically present such glycans. Therefore, we examined the therapeutic efficacy and toxicity of lectin drug conjugate therapy in CRC mouse xenograft models. The affinity of rBC2LCN for human CRC cell lines HT-29, LoVo, LS174T, and DLD-1 was assessed in vitro. The cytocidal efficacy of a lectin drug conjugate, rBC2LCN-38 kDa domain of pseudomonas exotoxin A (PE38) was evaluated by MTT assay. The therapeutic effects and toxicity for each CRC cell line-derived mouse xenograft model were compared between the intervention and control groups. LS174T and DLD-1 cell lines showed a strong affinity for rBC2LCN. In the xenograft model, the tumor volume in the rBC2LCN-PE38 group was significantly reduced compared with that using control treatment alone. However, the HT-29 cell line showed weak affinity and poor therapeutic efficacy. No significant toxicities or adverse responses were observed. In conclusion, we demonstrated that rBC2LCN lectin binds CRC cells and that rBC2LCN-PE38 significantly suppresses tumor growth in vivo. In addition, the efficacy of the drug conjugate correlated with its binding affinity for each CRC cell line. These results suggest that lectin drug conjugate therapy has potential as a novel targeted therapy for CRC cell surface glycans.


Assuntos
ADP Ribose Transferases/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Toxinas Bacterianas/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Exotoxinas/uso terapêutico , Imunoconjugados/uso terapêutico , Lectinas/uso terapêutico , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/efeitos adversos , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Toxinas Bacterianas/efeitos adversos , Burkholderia cenocepacia/química , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Portadores de Fármacos , Exotoxinas/efeitos adversos , Fucose/metabolismo , Fucosiltransferases/metabolismo , Células HT29 , Xenoenxertos , Humanos , Imunoconjugados/efeitos adversos , Técnicas In Vitro , Lectinas/isolamento & purificação , Lectinas/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/uso terapêutico , Carga Tumoral , Fatores de Virulência/efeitos adversos , Exotoxina A de Pseudomonas aeruginosa
6.
Oncologist ; 25(1): e170-e177, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31628266

RESUMO

Hairy cell leukemia (HCL) is an indolent B-cell malignancy characterized by high initial sensitivity to purine analog chemotherapy, minimal residual disease (MRD) frequently accompanying complete remission (CR), and relapses requiring additional treatment. Repeat chemotherapy shows decreasing efficacy and increasing toxicity with each course. Newer therapies targeting BRAF/MEK or Bruton's tyrosine kinase are effective but generally leave MRD. Rituximab has modest activity as a single agent and can achieve MRD-negative CR in combination with purine analogs, but there is significant toxicity from the chemotherapy. Moxetumomab pasudotox-tdfk (Moxe) is a biologic containing an antibody fragment (Fv) binding to CD22, attached to a portion of Pseudomonas exotoxin A. Binding to CD22 enables the toxin to enter and kill cells. Moxe is administered by 30-minute infusions on days 1, 3, and 5 of up to six cycles spaced 4 weeks apart. In phase I testing, 64% of 33 patients at the highest dose level achieved CR, most without MRD. Lack of MRD correlated with prolonged CR duration; of 11 MRD-negative CRs, 10 were still in CR after a median of 42 months of observation. In pivotal testing, 75% of 80 patients had a hematologic response, 41% with CR; 82% (27/33) of CRs were MRD-negative, and only 4 of the 27 MRD-negative patients relapsed during the follow-up period. Hemolytic uremic syndrome and capillary leak syndrome were each observed in 9% of patients, all reversible. In September 2018, the U.S. Food and Drug Administration approved Moxe for the treatment of relapsed/refractory HCL after ≥2 prior therapies. Moxe is undergoing further development in combination with rituximab. IMPLICATIONS FOR PRACTICE: Hairy cell leukemia (HCL) has effective treatments including purine analogs with and without rituximab, and oral inhibitors of BRAF, MEK and Bruton's tyrosine kinase (BTK). Despite these therapies, relapse occurs, and moxetumomab pasudotox has an important role in relapsed and refractory HCL because of its ability to achieve high rates of complete remissions (CRs) without chemotherapy; most of these CRs are without minimal residual disease (MRD). CR duration is enhanced in patients who achieve eradication of MRD. To improve the efficacy of this recombinant immunotoxin, a phase I trial is underway in combination with rituximab to reduce tumor burden and decrease immunogenicity.


Assuntos
Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Leucemia de Células Pilosas/tratamento farmacológico
7.
Blood ; 131(21): 2331-2334, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-29487070

RESUMO

Anti-CD22 moxetumomab pasudotox achieved 46% complete remissions (CRs) in previously reported phase 1 testing in relapsed/refractory hairy cell leukemia (HCL; n = 28). The importance of minimal residual disease (MRD) after CR in HCL is unknown. A 21-patient extension cohort received 50 µg/kg every other day for 3 doses in 4-week cycles. These patients plus 12 previously reported at this upper dose level received 143 cycles without dose-limiting toxicity. The combined 33-patient cohort achieved 64% CR and 88% overall response rates, with median CR duration of 42.4 months. Of 32 50-µg/kg patients evaluable for MRD by bone marrow aspirate flow cytometry (most stringent assessment), median CR duration was 13.5 (4.9-42.4) months in 9 MRD-positive CRs vs 42.1 (24.0-69.2) months in 11 MRD-negative CRs (P < .001). Among MRD-negative CRs, 10 patients had ongoing CR, 9 without MRD, at end of study. To our knowledge, moxetumomab pasudotox is the only nonchemotherapy regimen that can eliminate MRD in a significant percentage of HCL patients, to enhance CR duration. Repeated dosing, despite early neutralizing antibodies, increased active drug levels without detectable toxicity from immunogenicity. The activity and safety profiles of moxetumomab pasudotox support ongoing phase 3 testing in HCL. This trial was registered at www.clinicaltrials.gov as #NCT00586924.


Assuntos
Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Leucemia de Células Pilosas/tratamento farmacológico , Leucemia de Células Pilosas/patologia , Neoplasia Residual/tratamento farmacológico , Adulto , Idoso , Antineoplásicos/farmacologia , Toxinas Bacterianas/farmacologia , Ensaios Clínicos Fase I como Assunto , Monitoramento de Medicamentos , Exotoxinas/farmacologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
8.
Blood ; 130(14): 1620-1627, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28983018

RESUMO

Novel therapies are needed to overcome chemotherapy resistance for children with relapsed/refractory acute lymphoblastic leukemia (ALL). Moxetumomab pasudotox is a recombinant anti-CD22 immunotoxin. A multicenter phase 1 study was conducted to determine the maximum-tolerated cumulative dose (MTCD) and evaluate safety, activity, pharmacokinetics, and immunogenicity of moxetumomab pasudotox in children, adolescents, and young adults with ALL (N = 55). Moxetumomab pasudotox was administered as a 30-minute IV infusion at doses of 5 to 50 µg/kg every other day for 6 (cohorts A and B) or 10 (cohort C) doses in 21-day cycles. Cohorts B and C received dexamethasone prophylaxis against capillary leak syndrome (CLS). The most common treatment-related adverse events were reversible weight gain, hepatic transaminase elevation, and hypoalbuminemia. Dose-limiting CLS occurred in 2 of 4 patients receiving 30 µg/kg of moxetumomab pasudotox every other day for 6 doses. Incorporation of dexamethasone prevented further dose-limiting CLS. Six of 14 patients receiving 50 µg/kg of moxetumomab pasudotox for 10 doses developed hemolytic uremic syndrome (HUS), thrombotic microangiopathy (TMA), or HUS-like events, exceeding the MTCD. Treatment expansion at 40 µg/kg for 10 doses (n = 11) exceeded the MTCD because of 2 HUS/TMA/HUS-like events. Dose level 6B (ie, 50 µg/kg × 6 doses) was the MTCD, selected as the recommended phase 2 dose. Among 47 evaluable patients, an objective response rate of 32% was observed, including 11 (23%) composite complete responses, 5 of which were minimal residual disease negative by flow cytometry. Moxetumomab pasudotox showed a manageable safety profile and evidence of activity in relapsed or refractory childhood ALL. This trial was registered at www.clinicaltrials.gov as #NCT00659425.


Assuntos
Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Imunotoxinas/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Adolescente , Adulto , Toxinas Bacterianas/efeitos adversos , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/farmacocinética , Síndrome de Vazamento Capilar/prevenção & controle , Criança , Pré-Escolar , Dexametasona/uso terapêutico , Exotoxinas/efeitos adversos , Exotoxinas/imunologia , Exotoxinas/farmacocinética , Feminino , Glucocorticoides/uso terapêutico , Síndrome Hemolítico-Urêmica/induzido quimicamente , Humanos , Hipoalbuminemia/induzido quimicamente , Imunotoxinas/efeitos adversos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Lactente , Masculino , Dose Máxima Tolerável , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Microangiopatias Trombóticas/induzido quimicamente , Aumento de Peso/efeitos dos fármacos , Adulto Jovem
9.
J Oncol Pharm Pract ; 25(6): 1467-1472, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30917739

RESUMO

Hairy cell leukemia is a rare indolent B-cell lymphoid malignancy. Durable remission can be obtained with purine analogues, but relapse is inevitable, and effective treatment options may be limited. Moxetumomab pasudotox is a recombinant CD22-targeting immunotoxin that has recently been approved by the United States Food and Drug Administration for the treatment of relapsed or refractory hairy cell leukemia. Approval was based on a pivotal phase III study in this unique patient population. Rationale for use, clinical trial data, and current treatment recommendations are detailed. Common adverse effects are reviewed, and management strategies for select adverse effects are suggested. Implications for contemporary practitioners are also provided, as use of this novel agent is likely to increase as follow-up studies are reported.


Assuntos
Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Leucemia de Células Pilosas/tratamento farmacológico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico , Antineoplásicos/efeitos adversos , Antineoplásicos/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/patologia , Toxinas Bacterianas/efeitos adversos , Toxinas Bacterianas/metabolismo , Ensaios Clínicos Fase I como Assunto/métodos , Ensaios Clínicos Fase III como Assunto/métodos , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Exotoxinas/efeitos adversos , Exotoxinas/metabolismo , Seguimentos , Humanos , Hipotensão/induzido quimicamente , Leucemia de Células Pilosas/metabolismo , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Resultado do Tratamento
10.
World J Microbiol Biotechnol ; 35(6): 94, 2019 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-31187291

RESUMO

Pseudomonas aeruginosa is the major infectious agent of concern for cystic fibrosis (CF) patients. Therefore, it is necessary to develop appropriate strategies for preventing colonization by this bacterium and/or neutralizing virulence factors. In this study, we formulated the encapsulation of exotoxin A into PLGA nanoparticles. The biological activities of the nanovaccine candidate were also characterized. Based on the results, ETA-PLGA can act as a suitable immunogen to stimulate the humoral and cellular immune response. The antibodies raised against ETA-PLGA significantly decreased bacterial titer in the spleens of the immunized mice after challenge with PAO1 strain, compared to the control groups. The encapsulation of PLGA into ETA led to a significantly higher production of INF-γ, TNF-α, IL-4, and IL-17A cytokine responses compared to the ETA group. ETA-PLGA enhanced IgG responses in immunized mice compared to ETA antigen. We concluded that encapsulation of Pseudomonas aeruginosa ETA to PLGA nanoparticles can increase its functional activity by decreasing the bacterial dissemination.


Assuntos
ADP Ribose Transferases/imunologia , Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Imunização , Nanoconjugados , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/imunologia , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/patogenicidade , Vacinas Conjugadas , Fatores de Virulência/imunologia , ADP Ribose Transferases/uso terapêutico , Animais , Toxinas Bacterianas/uso terapêutico , Citocinas/metabolismo , Modelos Animais de Doenças , Exotoxinas/uso terapêutico , Feminino , Imunidade Celular , Imunidade Humoral , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/uso terapêutico , Infecções por Pseudomonas/imunologia , Baço/imunologia , Baço/microbiologia , Fatores de Virulência/uso terapêutico , Exotoxina A de Pseudomonas aeruginosa
11.
Cell Immunol ; 313: 59-66, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28087047

RESUMO

LMB-2, is a potent recombinant immunotoxin (RIT) that is composed of scFv antibody that targets CD25 (Tac) and a toxin fragment (PE38). It is used to treat T cell leukemias and lymphomas. To make LMB-2 less immunogenic, we introduced a large deletion in domain II and six point mutations in domain III that were previously shown to reduce T cell activation in other RITs. We found that unlike other RITs, deletion of domain II from LMB-2 severely compromised its activity. Rather than deletion, we identified T cell epitopes in domain II and used alanine substitutions to identify point mutations that diminished those epitopes. The novel RIT, LMB-142 contains a 38kDa toxin and nine point mutations that diminished T cell response to the corresponding peptides by an average of 75%. LMB-142 has good cytotoxic activity and has lower nonspecific toxicity in mice. LMB-142 should be more efficient in cancer therapy because more treatment cycles can be given.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Imunotoxinas/uso terapêutico , Leucemia de Células T/terapia , Pseudomonas/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais/genética , Toxinas Bacterianas/genética , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Desenho de Fármacos , ELISPOT , Epitopos de Linfócito T/genética , Exotoxinas/genética , Exotoxinas/uso terapêutico , Feminino , Engenharia Genética , Humanos , Imunotoxinas/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Leucemia de Células T/imunologia , Ativação Linfocitária , Camundongos , Mutagênese Sítio-Dirigida , Mutação/genética
12.
Biochem Biophys Res Commun ; 475(1): 93-9, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27178207

RESUMO

Immunotoxins have efficient anti-tumor activity due to their extreme potency. However, dose-limiting off-target toxicity and immunogenicity are the critical barriers for these immunotoxins to be used in a clinical setting. In this study, we designed a Pseudomonas exotoxin A (PE)-based human epidermal growth factor receptor-2 (HER2)-specific immunotoxin HER2-PE25-X7 by deleting most of domain II and introducing seven point mutations into domain III of the PE38 toxin. The anti-cancer activity, off-target toxicity and immunogenicity of this immunotoxin were carefully evaluated in vitro and in vivo. This new construct maintained the therapeutic potency of the original PE38-based immunotoxin HER2-PE38, with a greatly reduced off-target toxicity and immunogenicity. To compare with HER2-PE38, which resulted in the death of most of the mice after a single dose of 1.0 mg/kg, the new construct was completely tolerated at a dose of 10 mg/kg by the mice and almost completely depleted the tumor after treatment with five doses of 5 mg/kg of the immunotoxin. This work demonstrates a potentially attractive therapeutic modality for HER2-specific cancer treatment.


Assuntos
ADP Ribose Transferases/uso terapêutico , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Imunotoxinas/uso terapêutico , Neoplasias/tratamento farmacológico , Receptor ErbB-2/imunologia , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/imunologia , Animais , Antineoplásicos/imunologia , Toxinas Bacterianas/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Exotoxinas/imunologia , Feminino , Humanos , Imunotoxinas/imunologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia , Pseudomonas/imunologia , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
13.
J Investig Dermatol Symp Proc ; 17(2): 19-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26551939

RESUMO

Alopecia areata is an autoimmune condition where activated, pro-inflammatory white blood cells (WBCs) attack the hair follicles, resulting in hair loss. Migration of these activated WBCs from the blood stream and into the follicle tissue requires interaction between the integrin, lymphocyte function-associated antigen-1 (LFA-1) on WBCs, and ICAM-1 on vascular endothelial cells. High levels of active LFA-1 are uniquely expressed on WBCs that are involved in autoimmune and inflammatory conditions. The natural biologic agent LtxA (Leukothera) preferentially targets and depletes disease activated and malignant WBCs by binding to active LFA-1. The experimental drug has demonstrated significant therapeutic efficacy against autoimmune/inflammatory conditions such as psoriasis and allergic asthma in mouse models for these diseases. In addition, when injected into rodents, rhesus macaques, and dogs, LtxA was demonstrated to be physiologically active, biologically specific, and extremely well-tolerated. LFA-1 is an attractive target for therapy because it is only normally present on WBCs and has been shown to be activated and overexpressed on WBCs that are responsible for autoimmune/inflammatory conditions.


Assuntos
Alopecia em Áreas/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Doenças Autoimunes/tratamento farmacológico , Exotoxinas/farmacologia , Leucócitos/efeitos dos fármacos , Alopecia em Áreas/imunologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Morte Celular/efeitos dos fármacos , Exotoxinas/uso terapêutico , Humanos
14.
Br J Haematol ; 166(5): 729-38, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24931452

RESUMO

Frequencies of human leucocyte antigens (HLA) were determined in 287 classic hairy cell leukaemia (HCL) patients. With respect to both population (n = 287) and allele (2n = 574) frequency respectively, the most common HLA class I and II antigens expressed were HLA-A*02 (49·1% and 28·6%), HLA-B*07 (21·3% and 11·1%), HLA-C*07 (46·7 and 28·2%), HLA-DQB1*03 (62·7% and 37·3%), HLA-DRB1*11 (30·0% and 16·0%) and HLA-DRB4*01 (45·3% and 29·6%). In comparing 6-14 databases of control Caucasians to 267 Caucasian HCL patients, only HLA-DRB1*11 was consistently over-represented in HCL, 31·1% of patients vs. 17-19·9% of controls (P = 0·0055 to <0·0001) and 16·5% of alleles vs. 6·5-12·3% of control alleles (P = 0·022 to <0·0001). HLA-DRB1*11 is a known risk factor for acquired thrombotic microangiopathy. Anti-CD22 recombinant immunotoxin BL22 in HCL was associated with a 12% incidence of completely reversible grade 3-4 haemolytic uraemic syndrome (HUS), mainly during the second or third retreatment cycle. Of 49 HCL patients receiving ≥2 cycles of BL22, 7 (14%) had HUS and HLA-DRB1*11 was expressed in 71% of 7 with HUS compared with only 21% of 42 without (P = 0·015). These data suggest that DBR1*11 may be a marker for increased susceptibility to HCL and, among HCL patients, could be a risk factor for BL22-induced HUS.


Assuntos
Cadeias HLA-DRB1/biossíntese , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/metabolismo , Leucemia de Células Pilosas/genética , Leucemia de Células Pilosas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Feminino , Frequência do Gene , Predisposição Genética para Doença , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/terapia , Humanos , Leucemia de Células Pilosas/imunologia , Leucemia de Células Pilosas/terapia , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Microangiopatias Trombóticas , Doadores de Tecidos , Adulto Jovem
15.
Neoplasma ; 61(4): 392-400, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25027740

RESUMO

MSH-PE38KDEL is a chimeric molecule composed of MSH, and fused to a truncated mutant form of Pseudomonas exotoxin (PE38KDEL). Our study aims to evaluate the specific cytotoxicity of recombinant immunotoxin MSH-PE38KDEL on melanoma cells A875 and B16 in vitro, as well as its inhibition of metastatic melanoma in vivo. MSH-PE38KDEL was expressed in Escherichia coli, and greater than 90% purity was obtained. The purified MSH-PE38KDEL was found to be selectively cytotoxic to MSH receptor-positive melanoma cells in vitro. The specific cytotoxicity of recombinant MSH-PE38KDEL to A875 and B16 was over 85% by cell viability assay; however, MSH-PE38KDEL had no cytotoxicity to the human 2BS cells. The anti-tumor activity of MSH-PE38KDEL was evaluated in mice with induced melanoma through intra-tumor or intravenous administration. The results showed that 90% melanoma growths were inhibited, and 40% of the tumors were disappeared completely. Histopathology results showed MSH-PE38KDEL can effectively inhibit intrahepatic metastasis. In conclusion, MSH-PE38KDEL had cytotoxic effects on MSH receptor-positive melanoma cells, and causes significant tumor growth inhibition. These results support a possible new approach for the treatment of melanoma.


Assuntos
ADP Ribose Transferases/uso terapêutico , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Imunotoxinas/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Hormônios Estimuladores de Melanócitos/imunologia , Melanoma Experimental/tratamento farmacológico , Proteínas Recombinantes de Fusão/uso terapêutico , Fatores de Virulência/uso terapêutico , Animais , Feminino , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Células Tumorais Cultivadas , Exotoxina A de Pseudomonas aeruginosa
16.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 6-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24240707

RESUMO

In order to develop more effective therapeutic vaccines against cancers with high-risk human papillomavirus (HPV) infection, it is crucial to enhance the immunogenicity, eliminate the oncogenicity of oncoproteins, and take a combination of E7- and E6-containing vaccines. It has been shown recently that PE(ΔIII)-E7-KDEL3 (E7), a fusion protein containing the HPV16 oncoprotein E7 and the translocation domain of Pseudomonas aeruginosa exotoxin A, is effective against TC-1 tumor cells inoculated in mice, therefore, we engineered PE(ΔIII)-E6-CRL-KDEL3 (E6), the de-oncogenic versions of the E7 and E6 fusion proteins [i.e. PE(ΔIII)-E7(d)-KDEL3, E7(d), and PE(ΔIII)-E6(d)-CRL-KDEL3, E6(d)] and tested the immunoefficacies of these fusion proteins as mono- and bivalent vaccines. Results indicated that the E7(d) get higher immunogenicity than its wild type and the E6 fusion proteins augmented the immunogenicity and antitumor effects of their E7 counterparts. Furthermore, the bivalent vaccine system E7(d) plus E6(d), in the presence of cisplatin, showed the best tumoristatic and tumoricidal effects against established tumors in vivo. Therefore, it can be concluded that this novel therapeutic vaccine system, upon further optimization, may shed new light on clinical management of HPV-related carcinomas.


Assuntos
Cisplatino/uso terapêutico , Proteínas de Ligação a DNA/uso terapêutico , Proteínas de Fusão Oncogênica/uso terapêutico , Proteínas Oncogênicas Virais/uso terapêutico , Proteínas E7 de Papillomavirus/uso terapêutico , Vacinas contra Papillomavirus/uso terapêutico , ADP Ribose Transferases/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Exotoxinas/uso terapêutico , Camundongos , Proteínas de Fusão Oncogênica/imunologia , Vacinas contra Papillomavirus/imunologia , Fatores de Virulência/uso terapêutico , Exotoxina A de Pseudomonas aeruginosa
17.
J Control Release ; 367: 167-183, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37562556

RESUMO

The tumor microenvironment is a barrier to breast cancer therapy. Cancer-associated fibroblast cells (CAFs) can support tumor proliferation, metastasis, and drug resistance by secreting various cytokines and growth factors. Abnormal angiogenesis provides sufficient nutrients for tumor proliferation. Considering that CAFs express the sigma receptor (which recognizes anisamide, AA), we developed a CAFs and breast cancer cells dual-targeting nano drug delivery system to transport the LightOn gene express system, a spatiotemporal controlled gene expression consisting of a light-sensitive transcription factor and a specific minimal promoter. We adopted RGD (Arg-Gly-Asp) to selectively bind to the αvß3 integrin on activated vascular endothelial cells and tumor cells. After the LightOn system has reached the tumor site, LightOn gene express system can spatiotemporal controllably express toxic Pseudomonas exotoxin An under blue light irradiation. The LightOn gene express system, combined with multifunctional nanoparticles, achieved high targeting delivery efficiency both in vitro and in vivo. It also displayed strong tumor and CAFs inhibition, anti-angiogenesis ability and anti-metastasis ability, with good safety. Moreover, it improved survival rate, survival time, and lung metastasis rate in a mouse breast cancer model. This study proves the efficacy of combining the LightOn system with targeted multifunctional nanoparticles in tumor and anti-metastatic therapy and provides new insights into tumor microenvironment regulation.


Assuntos
Nanopartículas Multifuncionais , Nanopartículas , Neoplasias , Camundongos , Animais , Células Endoteliais , Exotoxinas/genética , Exotoxinas/uso terapêutico , Regulação da Expressão Gênica , Transgenes , Linhagem Celular Tumoral , Microambiente Tumoral , Nanopartículas/uso terapêutico
18.
Int J Cancer ; 133(6): 1497-506, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23494569

RESUMO

Cytoreductive surgery and intraperitoneal (i.p.) chemotherapy constitute a curative treatment option in mucinous peritoneal surface malignancies of intestinal origin, but treatment outcome is highly variable and the search for novel therapies is warranted. Immunotoxins are attractive candidates for targeted therapy in the peritoneal cavity because of direct cytotoxicity, distinct mechanisms of action and tumor cell selectivity. The MOC31PE immunotoxin targets the tumor-associated adhesion protein EpCAM (Epithelial Cell Adhesion Molecule), and has been administered safely in early clinical trials. In our work, the efficacy of i.p. administration of MOC31PE alone and together with mitomycin C (MMC) was investigated in unique animal models of human mucinous peritoneal surface malignancies. In initial model validation experiments, clear differences in efficacy were demonstrated between MMC and oxaliplatin, favoring MMC in five investigated tumor models. Subsequently, MOC31PE and MMC were given as single i.p. injections alone and in combination. In the PMCA-2 model, moderate growth inhibition was obtained with both drugs, while the combination resulted in at least additive effects; whereas the PMP-2 model was highly sensitive to both drugs separately and in combination and intermediate sensitivity was found for the PMCA-3 model. Furthermore, results from ex vivo experiments on freshly obtained mucinous tumor tissue from animals and patients suggested that classic mechanisms of immunotoxin activity were involved, i.e., inhibition of protein synthesis and induction of apoptosis. The present results suggest that adding MOC31PE to MMC-based i.p. chemotherapy should be further explored for EpCAM-expressing peritoneal surface malignancies, and a phase I trial is in preparation.


Assuntos
ADP Ribose Transferases/uso terapêutico , Antígenos de Neoplasias/imunologia , Toxinas Bacterianas/uso terapêutico , Moléculas de Adesão Celular/imunologia , Exotoxinas/uso terapêutico , Imunotoxinas/uso terapêutico , Neoplasias Peritoneais/tratamento farmacológico , Fatores de Virulência/uso terapêutico , Animais , Modelos Animais de Doenças , Molécula de Adesão da Célula Epitelial , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Mitomicina/uso terapêutico , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina , Neoplasias Peritoneais/patologia , Exotoxina A de Pseudomonas aeruginosa
19.
Int J Cancer ; 132(10): 2339-48, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23115013

RESUMO

Our study demonstrates the glioma tumor antigen podoplanin to be present at very high levels (>90%) in both glioblastoma (D2159MG, D08-0308MG and D08-0493MG) and medulloblastoma (D283MED, D425MED and DAOY) xenografts and cell line. We constructed a novel recombinant single-chain antibody variable region fragment (scFv), NZ-1, specific for podoplanin from the NZ-1 hybridoma. NZ-1-scFv was then fused to Pseudomonas exotoxin A, carrying a C-terminal KDEL peptide (NZ-1-PE38KDEL). The immunotoxin (IT) was further stabilized by a disulfide (ds) bond between the heavy-chain and light-chain variable regions as the construct NZ-1-(scdsFv)-PE38KDEL. NZ-1-(scdsFv)-PE38KDEL exhibited significant reactivity to glioblastoma and medulloblastoma cells. The affinity of NZ-1-(scdsFv), NZ-1-(scdsFv)-PE38KDEL and NZ-1 antibody for podoplanin peptide was 2.1 × 10(-8) M, 8.0 × 10(-8) M and 3.9 × 10(-10) M, respectively. In a protein stability assay, NZ-1-(scdsFv)-PE38KDEL retained 33-98% of its activity, whereas that of NZ-1-PE38KDEL declined to 13% of its initial levels after incubation at 37°C for 3 days. In vitro cytotoxicity of the NZ-1-(scdsFv)-PE38KDEL was measured in cells isolated from glioblastoma xenografts, D2159MG, D08-0308MG and D08-0493MG, and in the medulloblastoma D283MED, D425MED and DOAY xenografts and cell line. The NZ-1-(scdsFv)-PE38KDEL IT was highly cytotoxic, with an 50% inhibitory concentration in the range of 1.6-29 ng/ml. Significantly, NZ-1-(scdsFv)-PE38KDEL demonstrated tumor growth delay, averaging 24 days (p < 0.001) and 21 days (p < 0.001) in D2159MG and D283MED in vivo tumor models, respectively. Crucially, in the D425MED intracranial tumor model, NZ-1-(scdsFv)-PE38KDEL caused a 41% increase in survival (p ≤ 0.001). In preclinical studies, NZ-1-(scdsFv)-PE38KDEL exhibited significant potential as a targeting agent for malignant brain tumors.


Assuntos
ADP Ribose Transferases/imunologia , Toxinas Bacterianas/imunologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Exotoxinas/imunologia , Glioblastoma/tratamento farmacológico , Glioblastoma/imunologia , Meduloblastoma/tratamento farmacológico , Meduloblastoma/imunologia , Glicoproteínas de Membrana/imunologia , Anticorpos de Cadeia Única/imunologia , Fatores de Virulência/imunologia , ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Linhagem Celular Tumoral , Exotoxinas/uso terapêutico , Feminino , Humanos , Masculino , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Fatores de Virulência/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA